Language selection

Search

Patent 2722278 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2722278
(54) English Title: SYSTEMS, METHODS AND COMPOSITIONS FOR OPTICAL STIMULATION OF TARGET CELLS
(54) French Title: SYSTEMES, PROCEDES ET COMPOSITIONS PERMETTANT LA STIMULATION OPTIQUE DE CELLULES CIBLES
Status: Deemed expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 15/29 (2006.01)
  • C07K 14/415 (2006.01)
  • C12N 13/00 (2006.01)
  • C12N 15/00 (2006.01)
  • C12N 15/63 (2006.01)
  • C12N 15/85 (2006.01)
(72) Inventors :
  • ZHANG, FENG (United States of America)
  • DEISSEROTH, KARL (United States of America)
  • GRADINARU, VIVIANA (United States of America)
(73) Owners :
  • THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY (United States of America)
(71) Applicants :
  • THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY (United States of America)
(74) Agent: SMART & BIGGAR IP AGENCY CO.
(74) Associate agent:
(45) Issued: 2020-03-31
(86) PCT Filing Date: 2009-04-08
(87) Open to Public Inspection: 2009-10-29
Examination requested: 2014-04-08
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2009/039949
(87) International Publication Number: WO2009/131837
(85) National Entry: 2010-10-22

(30) Application Priority Data:
Application No. Country/Territory Date
61/047,219 United States of America 2008-04-23

Abstracts

English Abstract



Methods, systems and devices are implemented in connection with light-
responsive ion channel molecules. One
such method is implemented using a light- activated ion channel molecule that
responds to a light stimulus. The method includes
engineering the light-activated ion channel molecule in a cell; and activating
the ion channel molecule, in response to light stimulus
that is provided to the ion channel molecule and that has properties that do
not activate a ChR2 ion channel, to allow ions to
pass through the light-activated ion channel molecule.


French Abstract

Linvention concerne des procédés, systèmes et dispositifs mis en uvre en relation avec les molécules des canaux ioniques sensibles à la lumière. Un tel procédé est mis en uvre à laide dune molécule de canal ionique activée par la lumière répondant à un stimulus lumineux. Le procédé comprend la génération de la molécule de canal ionique activée par la lumière dans une cellule; et lactivation de ladite molécule de canal ionique en réaction au stimulus lumineux appliqué à ladite molécule de canal ionique et possédant des propriétés nactivant pas le canal ionique ChR2, pour permettre aux ions de passer à travers ladite molécule de canal ionique.

Claims

Note: Claims are shown in the official language in which they were submitted.


52
Claims:
1. A mammalian cell genetically modified to express a light-responsive ion
channel protein from Volvox carteri, wherein the ion channel protein comprises
the amino acid
sequence set forth in SEQ ID NO:3.
2. The cell of claim 1, wherein the ion channel protein exhibits excitation
in a
range of from about 535 nm to about 589 nm.
3. The cell of claim 1 or 2, wherein the cell is a neuronal cell or a stern
cell.
4. The cell of claim 1, 2 or 3, wherein the ion channel protein is encoded
by a
nucleotide sequence operably linked to an alpha-CaMKII promoter.
5. The cell of any one of claims 1 to 4, wherein the ion channel protein is
encoded
by a nucleotide sequence that is codon optimized for expression in a mammalian
cell.
6. The cell of any one of claims 1 to 5, wherein the cell further expresses
an NpHR
halorhodopsin (NpHR) protein from Natronomonas pharaonis.
7. The cell of claim 6, wherein the NpHR protein is a light-responsive NpHR

protein.
8. The cell of any one of claims 1 to 7, wherein the cell further comprises
a light-
responsive ChR2 channelrhodopsin (ChR2) protein from Chlamydomonas
reinhardtii.
9. The cell of claim 8, wherein the ChR2 protein comprises the amino acid
sequence set forth in SEQ ID NO:2.
10. A population of cells comprising cells as defined in any one of claims
1 to 9.

53

11. A recombinant expression vector comprising a nucleotide sequence
encoding a
polypeptide comprising the amino acid sequence set forth in SEQ ID NO:3.
12. The recombinant expression vector of claim 11, wherein the nucleotide
sequence is codon optimized for expression in a mammalian cell.
13. The recombinant expression vector of claim 11 or 12, wherein the
nucleotide
sequence is operably linked to a promoter.
14. The recombinant expression vector of claim 13, wherein the promoter is
an
alpha-CaMKII promoter.
15. The recombinant expression vector of any one of claims 11 to 14,
wherein the
expression vector is a viral vector.
16. The recombinant expression vector of claim 15, wherein the viral vector
is a
lentiviral vector or an adenoassociated virus (AAV) vector.
17. An ex vivo method for modulating the activity of a cell engineered to
express a
Volvox carteri light-responsive ion channel protein, the method comprising
activating the light-
responsive ion channel protein with light, wherein the light-responsive ion
channel protein
comprises the amino acid sequence set forth in SEQ ID NO:3.
18. The method of claim 17, wherein the cell is a stern cell.
19. The method of claim 17, wherein the cell is a neuronal cell.
20. The method of claim 17, 18 or 19, wherein the light-responsive protein
is
encoded by a nucleotide sequence operably linked to an alpha-CaMKII promoter.

54

21. The method of any one of claims 17 to 20, wherein the light-responsive
protein
is activated by light having a wavelength in a range of from about 535 nm to
about 589 nm.
22. The method of any one of claims 17 to 21, wherein the light-responsive
protein
is encoded by a nucleotide sequence that is optimized for expression in a
mammalian cell.
23. The method of any one of claims 17 to 22, wherein the light-responsive
protein
is encoded by an expression vector.
24. The method of claim 23, wherein the expression vector is a viral
vector.
25. The method of claim 24, wherein the viral vector is a lentiviral vector
or an
adenoassociated virus (AAV) vector.
26. The method of any one of claims 17 to 25, wherein the cell is further
engineered
to express the ChR2 channelrhodopsin from Chlamydomonas reinhardtii.
27. The method of claim 26, wherein the ChR2 channelrhodopsin comprises the

amino acid sequence set forth in SEQ ID NO:2.
28. The method of claim 26 or 27, wherein the ChR2 channelrhodopsin is
activated
by light having a wavelength of 589 nm.
29. The method of any one of claims 17 to 28, wherein the cell is further
engineered
to express the NpHR halorhodopsin from Natronomonas pharaonis.
30. Use of a Volvox carteri light-responsive ion channel protein comprising
the
amino acid sequence set forth in SEQ ID NO:3 for modulating the activity of a
cell engineered
to express the light-responsive ion channel protein.
31. The use of claim 30, wherein the cell is a stem cell.

55

32. The use of claim 30, wherein the cell is a neuronal cell.
33. The use of claim 32. wherein the light-responsive protein is encoded by
a
nucleotide sequence operably linked to an alpha-CaMKII promoter.
34. The use of any one of claims 30 to 33, wherein the light-responsive
protein is
activated by light having a wavelength in a range of from about 535 mn to
about 589 mu.
35. The use of any one of claims 30 to 34, wherein the light-responsive
protein is
encoded by a nucleotide sequence that is optimized for expression in a
mammalian cell.
36. The use of any one of claims 30 to 35, wherein the light-responsive
protein is
encoded by an expression vector.
37. The use of claim 36, wherein the expression vector is a viral vector.
38. The use of claim 37, wherein the viral vector is a lentiviral vector or
an
adenoassociated virus (AAV) vector.
39. The use of any one of claims 30 to 38, wherein the cell is further
engineered to
express the ChR2 channelrhodopsin from Chlamydomonas reinhardtii.
40. The use of claim 39, wherein the ChR2 channelrhodopsin comprises the
amino
acid sequence set forth in SEQ ID NO:2.
41. The use of claim 39 or 40, wherein the ChR2 channelrhodopsin is
activated by
light having a wavelength of 589 nm.
42. The use of any one of claims 30 to 41, wherein the cell is further
engineered to
express the NpHR halorhodopsin from Natronomonas pharaonis.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02722278 2014-04-08
1
SYSTEMS, METHODS AND COMPOSITIONS
FOR OPTICAL STIMULATION OF TARGET CELLS
Sequence Listing
The description contains a sequence listing in electronic form in ASCII text
format. A copy
of the sequence listing in electronic from is available from the Canadian
Intellectual Property
Office.
Overview
The stimulation of various cells of the body has been used to produce a number
of
beneficial effects. One method of stimulation involves the use of electrodes
to introduce an
externally generated signal into cells. One problem faced by electrode-based
brain stimulation
techniques is the distributed nature of neurons responsible for a given mental
process. Conversely,
different types of neurons reside close to one another such that only certain
cells in a given region
of the brain are activated while performing a specific task. Alternatively
stated, not only do
heterogeneous nerve tracts move in parallel through tight spatial confines,
but the cell bodies
themselves may exist in mixed, sparsely embedded configurations. This
distributed manner of
processing seems to defy the best attempts to understand canonical order
within the CNS, and
makes neuromodulation a difficult therapeutic endeavor. This architecture of
the brain poses a
problem for electrode-based stimulation because electrodes are relatively
indiscriminate with
regards to the underlying physiology of the neurons that they stimulate.
Instead, physical proximity
of the electrode poles to the neuron is often the single largest determining
factor as to which
neurons will be stimulated. Accordingly, it is generally not feasible to
absolutely restrict
stimulation to a single class of neurons using electrodes.
Another issue with the use of electrodes for stimulation is that because
electrode placement
dictates which neurons will be stimulated, mechanical stability is frequently
inadequate, and results
in lead migration of the electrodes from the targeted area. Moreover, after a
period of time within
the body, electrode leads frequently become encapsulated with glial cells,
raising the effective
electrical resistance of the electrodes, and hence the electrical power
delivery required to reach

CA 02722278 2015-12-02
CA2722278
2
targeted cells. Compensatory increases in voltage, frequency or pulse width,
however, may spread
the electrical current and increase the unintended stimulation of additional
cells.
Another method of stimulus uses photosensitive bio-molecular structures to
stimulate target
cells in response to light. For instance, light activated proteins or
molecules can be used to control the
flow of ions through cell membranes. By facilitating or inhibiting the flow of
positive or negative
ions through cell membranes, the cell can be briefly depolarized, depolarized
and maintained in that
state, or hyperpolarized. Neurons are an example of a type of cell that uses
the electrical currents
created by depolarization to generate communication signals (i.e., nerve
impulses). Other electrically
excitable cells include skeletal muscle, cardiac muscle, and endocrine cells.
Neurons use rapid
depolarization to transmit signals throughout the body and for various
purposes, such as motor
control (e.g., muscle contractions), sensory responses (e.g., touch, hearing,
and other senses) and
computational functions (e.g., brain functions). Thus, the control of the
depolarization of cells can be
beneficial for a number of different purposes, including (but not limited to)
psychological therapy,
muscle control and sensory functions.
Summary
Aspects of this disclosure are directed to photosensitive bio-molecular
structures and related
methods. Such subject matter is exemplified in a number of implementations and
applications, some
of which are summarized below.
According to one example embodiment, an implantable arrangement is implemented
having a
.. light-generation device for generating light. The arrangement also has a
biological portion that
modifies target cells for stimulation in response to light generated by the
light-generation means in
vivo.
According to another example embodiment, target cells are stimulated using an
implantable
arrangement. The arrangement includes an electrical light-generation means for
generating light and
a biological portion. The biological portion has a photosensitive bio-
molecular arrangement that
responds to the generated light by stimulating target cells in vivo.
Stimulation may be manifested as
either up-regulation, or down-regulation of activity at the target.
In accordance with one aspect, the present disclosure provides a mammalian
cell expressing a
light-responsive VChR1 ion channel from Volvox carteri. In one embodiment, the
VChR1 ion
channel exhibits excitation in a range of from about 535 nm to about 589 nm.
In another

CA 02722278 2015-12-02
CA2722278
2a
embodiment, the VChR1 channel comprises the amino acid sequence set forth in
SEQ ID NO: 3. In
another embodiment, the cell is a neuronal cell or a stem cell. In another
embodiment, the VChR1
ion channel is encoded by a nucleotide sequence operably linked to an alpha-
CaMKII promoter. In
another embodiment, the VChR1 ion channel is encoded by a nucleotide sequence
that is codon
optimized for expression in a mammalian cell. In another embodiment, the cell
also expresses an
NpHR protein.
In accordance with another aspect, the present disclosure provides a
population of cells
comprising cells as defined herein. In one embodiment, the population of cells
further comprises
cells containing a light-responsive ChR2 protein from Chlamydomonas
reinhardtii. In another
embodiment, the ChR2 protein comprises the amino acid sequence set forth in
SEQ ID NO:2. In
another embodiment, the population of cells further comprises cells containing
a light-responsive
NpHR protein from Natronomonas pharaonis.
In accordance with another aspect, the present disclosure provides a
polynucleotide
encoding a light-responsive VChR1 protein from Volvox carteri for use in the
preparation of a
cell as defined herein. In one embodiment, the polynucleotide comprises a
nucleotide sequence
encoding the amino acid sequence set forth in SEQ ID NO:3. In another
embodiment, the
nucleotide sequence is codon optimized for expression in a mammalian cell. In
another
embodiment, the nucleotide sequence is operably linked to a promoter. In one
embodiment, the
promoter is an alpha-CaMKII promoter. In another embodiment, the
polynucleotide is in an
expression vector. In one embodiment, the expression vector is a viral vector.
In one
embodiment, the viral vector is a lentiviral vector or an adenoassociated
virus (AAV) vector.
In accordance with another aspect, the present disclosure provides a method
for
modulating the activity of a cell engineered to express a Volvox carteri light-
responsive ion
channel protein (VChR1), the method comprising activating the light-responsive
VChR1 protein
with light. In one embodiment, the cell may be a stem cell. In another
embodiment, the cell may
be a neuronal cell. In another embodiment, the light-responsive VChR1 protein
is encoded by a
nucleotide sequence operably linked to an alpha-CaMKII promoter. In another
embodiment, the
light-responsive VChR1 protein is activated by light having a wavelength in a
range of from
about 535 nm to about 589 nm. In another embodiment, the light-responsive
VChR1 protein
comprises the amino acid sequence set forth in SEQ ID NO:3, In another
embodiment, the light-
responsive VChR1 protein is encoded by a nucleotide sequence that is optimized
for expression in a

CA 02722278 2016-12-02
CA 2722278
2b
mammalian cell. In one embodiment, the light-responsive VChR1 protein is
encoded by an
expression vector. In one embodiment, the expression vector may be a viral
vector. In one
embodiment, the viral vector is a lentiviral vector or an adenoassociated
virus (AAV) vector. In
another embodiment, the cell is further engineered to express a light-
responsive ChR2 protein
.. from Chlamydomonas reinhardtii. In another embodiment, the ChR2 protein
comprises the
amino acid sequence set forth in SEQ ID NO:2. In another embodiment, the cell
is further
engineered to express a light-responsive NpHR protein from Natronomonas
pharaonis.
The claimed invention relates to a mammalian cell genetically modified to
express a
light-responsive ion channel protein from Volvox carteri, wherein the ion
channel protein
.. comprises the amino acid sequence set forth in SEQ ID NO:3. Also claimed is
a population of
cells comprising such genetically modified mammalian cells. Also claimed is a
recombinant
expression vector comprising a nucleotide sequence encoding a polypeptide
comprising the
amino acid sequence set forth in SEQ ID NO:3. Also claimed is a method for
modulating the
activity of a cell engineered to express a Volvox carteri light-responsive ion
channel protein, the
.. method comprising activating the light-responsive ion channel protein with
light, wherein the
light-responsive ion channel protein comprises the amino acid sequence set
forth in SEQ ID
NO:3.

CA 02722278 2015-12-02
3
According to another example embodiment, an
implantable device delivers gene transfer vector, such as a virus, which
induces
expression of photosensitive bio-molecular membrane proteins. The device has a
light
generator, responsive to (for example, charged by or triggered by) an external
signal, to
generate light and a biological arrangement that includes the photosensitive
bio-molecular
protein that responds to the generated light by interacting with target cells
in vivo. In this
manner, the electronic portions of the device may be used to optically
stimulate target
cells. Stimulation may be manifested as either up-regulation (e.g., increased
neuronal
firing activity), or down-regulation (e.g., neuronal hyper-polarization, or
alternatively,
chronic depolarization) of activity at the target.
According to another example embodiment , , a method is
implemented for stimulating target cells using photosensitive proteins that
bind with the
target cells. The method includes a step of implanting the photosensitive
proteins and a
light generating device near the target cells. The light generating device is
activated and
the photosensitive protein stimulates the target cells in response to the
generated light.
Applications include those associated with any population of electrically-
excitable
cells, including neurons, skeletal, cardiac, smooth muscle cells, and insulin-
secreting
pancreatic beta cells. Major diseases with altered excitation-effector
coupling include
heart failure, muscular dystrophies, diabetes, pain, cerebral palsy,
paralysis, depression,
and schizophrenia.
According to other example embodiments, methods for
generating an excitation neuron-current flow involve, in a neuron, engineering
a protein
that responds to light by producing an excitation current to encourage
depolarization of
the neuron. In one such method, the protein is derived from Volvox carteri.
The above summary is not intended to describe each
illustrated embodiment or every implementation The figures and
detailed description that follow more particularly exemplify these
embodiments.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
4
Brief Description of the Drawings
The invention may be more completely understood in consideration of the
detailed
description of various embodiments of the invention that follows in connection
with the
accompanying drawings, in which:
FIG. 1A shows a pheroidal alga Volvox carteri, consistent with an example
embodiment of the present invention;
FIG. 1B shows all-trans retinal Schiff base and related protein sequences,
consistent with an example embodiment of the present invention;
FIG. 1C evoked photocurrents relative to light intensity, consistent with an
example embodiment of the present invention;
FIG. 1D shows an inwardly rectifying current-voltage relationship, consistent
with
an example embodiment of the present invention;
FIG. lE shows membrane currents relative to specific ions, consistent with an
example embodiment of the present invention;
FIG. 1F shows activation percentage relative to optical wavelength, consistent
with an example embodiment of the present invention;
FIG. 2A shows neurons expressing VChRl-EYFP and exhibiting membrane-
localized EYFP fluorescence, consistent with an example embodiment of the
present
invention;
FIG. 2B shows VChRl-EYFP neurons photocurrents when illuminated with
531nm and 589nm light, consistent with an example embodiment of the present
invention;
FIG. 2C shows whole-cell inward currents for 531nm and 589nm light, consistent

with an example embodiment of the present invention;
FIG. 2D shows twenty 5ms light pulses delivered to VChRl-EYFP neurons in
.. current clamp at various frequencies, consistent with an example embodiment
of the
present invention;
FIG. 2E shows the percentages of successful spikes at various frequencies,
consistent with an example embodiment of the present invention;
FIG. 2F shows that increasing frequencies of light pulses delivered increased
steady-state depolarization, consistent with an example embodiment of the
present
invention;
FIG. 2G shows the membrane resistance, consistent with an example embodiment
of the present invention;

CA 02722278 2015-12-02
FIG. 2H shows resting membrane potential, consistent with an example
embodiment of the present invention;
FIG. 3A shows voltage responses to optical stimulation at different
wavelengths,
consistent with an example embodiment of the present invention;
5 FIG. 3B shows a percentage of successful spikes for optical stimulation
at
different wavelengths and intensities, consistent with an example embodiment
of the
present invention;
FIG. 3C shows a percentage of successful spikes for optical stimulation at
different wavelengths and intensities, consistent with an example embodiment
of the
present invention;
FIG. 4A-D shows direct optical inhibition of local subthalamic nucleus (STN)
neurons;
FIG, 5A-C shows targeting astroglia within the STN;
FIG. 6A-C shows optical depolarization of STN neurons at different
frequencies;
FIG. 7A-C shows quantification of the tissue volume recruited by optical
intervention;
FIG. 8A-C shows selective optical control of afferent fibers in the STN;
FIG. 9A-D shows selective optical stimulation of layer V neurons in anterior
primary motor cortex;
FIG. 10A-C shows substantia nigra lesion and cannula track;
FIG. 11A-C shows an additional histological characterization;
FIG. 12A-D shows additional behavioral results;
FIG. 13A-D shows additional electrophysiological results;
FIG. 14A-D shows high-temporal resolution optrode traces;
FIG. 15A-C shows latency of M1 response to optical stimulation of STN; and
FIG. 16A-F shows changes in frequency characteristics of neuronal activity
produced by optical stimulation
While the invention is amenable to various modifications and alternative
forms,
specifics thereof have been shown by way of example in the drawings and will
be
described in detail. it should be understood, however, that the intention is
not to limit the
invention to the particular embodiments described.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
6
Detailed Description
The present invention is believed to be useful for facilitating practical
applications
of a variety of photosensitive bio-molecular structures, and the invention has
been found
to be particularly suited for use in arrangements and methods dealing with
cellular
membrane voltage control and stimulation. While the present invention is not
necessarily
limited to such applications, various aspects of the invention may be
appreciated through
a discussion of various examples using this context.
Consistent with one example embodiment of the present invention, a light-
responsive protein/molecule is engineered in a cell. The protein affects a
flow of ions
across the cell membrane in response to light. This change in ion flow creates
a
corresponding change in the electrical properties of the cells including, for
example, the
voltage and current flow across the cell membrane. In one instance, the
protein functions
in vivo using an endogenous cofactor to modify ion flow across the cell
membrane. In
another instance, the protein changes the voltage across the cell membrane so
as to
dissuade action potential firing in the cell. In yet another instance, the
protein is capable
of changing the electrical properties of the cell within several milliseconds
of the light
being introduced.
Consistent with a more specific example embodiment of the present invention, a

protein, herein identified as VChR1, from Volvox carteri is used for
temporally-precise
optical control of neural activity. VChR1 allows for selective excitation of
single action
potentials including the generation of rapid spike trains and sustained
blockade of spiking
over many minutes. The action spectrum of VChR1 is strongly red-shifted
relative to
ChR2 but operates at similar light power, and functions in mammals without
exogenous
cofactors. In one instance, VChR1 can be co-expressed with and NpHR and/or
ChR2 in
the target cells. Likewise, VChR1, NpHR and ChR2 can be targeted to C. elegans
muscle
and cholinergic motoneurons to control locomotion bidirectionally. In this
regard,
VChR1, NpHR and ChR2 form an optogenetic system for multimodal, high-speed,
genetically-targeted, all-optical interrogation of living neural circuits.
Embodiments of the present invention are directed to the VChR1 protein.
Various
embodiments are directed toward a plasmid that contains the DNA or nucleotide
sequence
that expresses the VChR1 protein. Yet other embodiments are directed toward an

expression vector for expression of the VChR1 protein. A non-exclusive list of

CA 02722278 2010-10-22
WO 2009/131837 7 PCT/US2009/039949
expression vectors includes bacterial, viral and plant plasmids. Another
embodiment of
the present invention is directed to heterologous cells that contain the VChR1
protein.
Aspects of the present invention are directed toward variations of the
specific
embodiment of VChR1 disclosed in FIG. 1B. One such aspect includes mutations
of the
protein. These mutations may, for example, target portions of the VChR1
protein that
shift or otherwise change the wavelength of light that activates the protein.
Fast light-activated microbial proteins adapted for neuroscience, including
the
channelrhodopsin ChR2 and the halorhodopsin NpHR, permit millisecond-precision

optical control of genetically-defined cell types within intact neural tissue.
Since ChR2 is
a blue light-driven cation channel that can activate neurons, and NpHR is a
yellow light-
driven chloride pump that can inhibit neurons, the combination of these two
proteins
allows independent neural excitation and inhibition in the same preparation. A
third
major optogenetic tool, namely, a second cation channel with action spectrum
significantly shifted relative to ChR2, would allow experimental testing of
the differential
.. contribution or interaction of two distinct cell types in circuit
computation or behavior.
One ChR2-related sequence from the spheroidal alga Volvox carter! (FIG. 1A)
has
been described, but the absorption spectrum of the protein and the photocycle
dynamics
are virtually identical to those of ChR2. A second Vo/vox ChR more related to
ChR1
from Chlamydomonas reinhardtii (FIG. 1B) was discovered. This new protein and
.. variants thereof are herein referred to as VChR1.
In an experimental test, VChR1 was expressed in Xenopus oocytes and HEK293
cells, and found to evoke photocurrents were similar to those of ChR1 from
Chlamydomonas. The photocurrents were graded with light intensity, and
displayed
inactivation from a fast peak toward a slightly reduced stationary plateau
(FIG. 1C). The
.. peak appeared preferentially at light of relatively high intensity, likely
attributable to
increased accumulation of an expected late non-conducting photocycle
intermediate
(FIG. 1C), and as light intensity approached saturation the evoked current
displayed a
characteristic minimum before steady-state is reached. VChR1 exhibited an
inwardly
rectifying current-voltage relationship (FIG. 1D) and under neuronal
physiological
.. conditions conducted chiefly Na+ but also H+, K+, and Ca2+ (FIG. 1E).
Primary-structural differences between VChR1 and the Chlamydomonas ChRs
were identified to allow for prediction of the altered absorption properties
(FIG. 1B,
depicting SEQ. ID. NOs. 001 (ChR2), 002 (ChR1) and 003 (VChR1), with primary-

CA 02722278 2010-10-22
WO 2009/131837 8
PCT/US2009/039949
structural differences highlighted). First, based on previous calculations of
the
electrostatic potential of bacteriorhodopsin (BR, absorption maximum at 570
nm) and
sensory rhodopsin II (SRII, absorption maximum at 500 nm) and on additional
quantum
mechanical-molecular mechanical calculations (QM/MM), the counterion complex
of the
cofactor all-trans retinal Schiff base (RSB; FIG. 1B) will likely be most
critical for color
tuning, photoisomerization and photocycle dynamics. Based on homology with
other
microbial opsin genes for which the 3D structure is known, the counterion
complex in
ChR2 should be defined by R120, E123, and D253. However, these residues are
fully
conserved in both ChR1 and VChR1 (FIG. 1B, highlighted columns 104). Second,
theoretical calculations in line with previous mutational experiments predict
that three
residues of the RSB binding pocket could significantly contribute to
absorption
differences among microbial rhodopsin proteins. These amino acids are G181,
L182, and
S256 in ChR2 (FIG. 1B sequence, highlighted columns 106); the former two are
expected
to be located near the RSB p-ionone ring (FIG. 1B structure, 108) and may, in
conjunction with C183, determine absorbance spectrum, while S256 is instead
likely
adjacent to the protonated nitrogen of the RSB (FIG. 1G structure, 110). In
VChR1 the 13-
ionone ring end of the RSB is expected to be more polar than in ChR1 and ChR2,
since
the two positions 181 and 183 have been substituted with a polar Ser, while
conversely
the RSB nitrogen environment is actually less polar with an Ala at position
256. The
combination of these three exchanges at positions 181, 183, and 256 resulting
in an
expectation of a redistribution of positive charge along the RSB polyene
system and a
substantial redshift, likely by more than 40 nm, in VChR1 compared to ChR2.
Three other amino acids H114, E235, and E245 (FIG. 1B, highlighted columns
102) are expected to modulate the RSB charge distribution by long-range
coupling, and
here the H1 14N exchange in both VChR1 and ChR1 is further predicted to
increase the
RSB potential at the 13-ionone end. The 495 nm absorbance maximum of ChR1
(which
does not express well in neurons), is indeed slightly red-shifted from that of
ChR2, but
the combination of many significant changes in VChR1 predicted a robust
wavelength
shift on a scale useful for defining a new class of tool for neuroscience.
To initially probe the wavelength-dependence, VChR1 -expressing oocytes were
excited using 10 ns laser flashes across a range of wavelengths, to allow
delineation of a
markedly red-shifted action spectrum that revealed a maximum at ¨535 nm and a
small
shoulder at lower wavelengths consistent with a second isoform peaking at 505
nm

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
9
(FIG. 1F). A lentivirus carrying the alpha-CaMKII promoter to drive strong
protein
expression was constructed to test the function of VChR1 in neurons. To
visualize
VChR1 expression, the seven transmembrane domains of VChR1 (residues 1-300,
based
on homology with the first 315 residues of ChR2) were in-frame fused to the
amino-
terminus of yellow fluorescent protein (VChR1-EYFP). Neurons expressing VChRl-
EYFP exhibited clearly membrane-localized EYFP fluorescence similar to that
reported
previously for ChR2-EYFP (FIG. 2A), with expression level slightly weaker
compared
with ChR2-EYFP using the same lentiviral alpha-CaMKII expression vector.
Nevertheless, VChR1-EYFP neurons exhibited strong photocurrents when
illuminated
with 531 nm and even 589 nm light (FIG. 2B). Mean whole-cell inward currents
were
208.8122.3 pA (mean s.e.m. reported unless otherwise stated, n=20) and
177.6124.7 pA
(n=10) when illuminated with 15mW/mm2 of 531 nm light and 13.8 mW/mm2 of 589
nm
light at the sample, respectively (FIG. 2C). Apparent time constants for the
rise of the
photocurrent were faster when closer to the wavelength of maximum activation
due to the
.. shift in absorption coefficient, with corresponding values of T53 l_on =
2.810.3 ms and
T589_on = 8.010.7 ms (n = 11 for 531 nm and n = 10 for 589 nm). The
corresponding
decay time constants were -631 off = 133.4111.7 ms (n = 11) and T589 off =
135.719.8
ms (n = 10).
The frequency dependence of VChR1 in evoking spikes was explored using trains
of twenty 5 ms light pulses at 531 nm or 589 nm delivered to VChR1-EYFP
neurons in
current clamp (exemplar traces from 589 nm excitation in FIG. 2D). At up to 10
Hz,
more than 90% of tested cells fired 100% of the action potentials in the train
at either
wavelength, and at 20 Hz cells typically fired in response to ¨65% of light
pulses
(FIG. 2E). In these strongly expressing cells, reliable spiking could be
driven up to 30 Hz
(FIG. 2D; pyramidal neurons in culture typically cannot follow 50 Hz or beyond
in
response to either current injection or ChR2 photostimulation), and at 531 nm,
doublets of
spikes were occasionally evoked for each light pulse, most likely due to the
slower Toff
decay constant of 133 ms compared to 12 ms for ChR210. As with ChR2, VChR1
could
also trigger EPSP-like subthreshold depolarizations with lower stimulation
light
intensities. Delivery of light pulse barrages evoked typical summation of the
subthreshold membrane voltage changes, with increasing frequencies of light
pulses
delivering increased steady-state depolarization (FIG. 2F).

CA 02722278 2010-10-22
WO 2009/131837
PCT/US2009/039949
To test for possible effects on membrane integrity, the membrane resistance
and
resting membrane potential were compared (FIGs. 2G and 2H) by whole-cell patch
clamp,
among 1) VChR1-EYFPexpressing, 2) non-transduced, and 3) VChR1-EYFP-expressing

neurons first patch-clamped 24 hr after exposure to a typical light pulse
protocol (1 s of
5 20 Hz, 5 ms light flashes, once per minute, for 10 minutes). All cells
recorded exhibited
comparable values, suggesting that VChR1-EYFP expression did not significantly
alter
membrane electrical properties. Subcellular distribution appeared similar to
that of ChR2,
with strong membrane localization, and VChR1 was well tolerated by these
neurons.
Moreover, as with ChR2 and NpHR, no all-trans retinal supplementation was
needed with
10 .. after VChR1 transduction in neurons; these genes all encode microbial
opsins, which
require incorporation of all-trans retinal to form the RSB and become
functional
rhodopsins, but vertebrate neurons consistently have been found to convert
expressed
opsins into functional proteins without supplementation of chemical cofactors.
Testing was performed as to whether the pronounced spectral separation between
.. ChR2 and VChR1 activation would be sufficient to enable separable
activation using two
different wavelengths of light. Based upon the action spectra (FIG. 1F), 406nm
and
589nm were selected as likely optimal excitation wavelengths to probe
separable
activation of ChR2 and VChR1. For neurons expressing either ChR2 or VChR1,
testing
was performed for evoked action potentials in response to trains of twenty 5ms
light
pulses (406nm and 589nm) delivered at 5Hz. Each wavelength was tested at
several
different light intensities to determine parameters that maximize ChR2
activation while
minimizing VChR1 activation at 406nm, and vice versa. It was discovered that
ChR2 and
VChR1 neurons can be separately activated by 406nm and 589nm light
respectively
(FIG. 3A). In fact, no ChR2 neurons fired action potentials when illuminated
with 589nm
light pulses since the absorption is practically zero at this wavelength,
whereas VChR1
neurons fired reliably at this wavelength. Conversely all ChR2 neurons fired
20 action
potentials when illuminated with 406nm light, at all three light intensities
(n=10, FIG. 3B).
While VChR1 cells were capable of firing occasional action potentials in
response to
406nm flashes (generally, all rhodopsins exhibit some absorption at this
wavelength due
to transition to the second electronic state, SO -> S2 transition), the
percentage of spikes
could be reduced to 13 9% when the 406nm light intensity was reduced to 1.2
mW/mm2
(n=10, FIG. 3C), an intensity which continued to reliably and robustly drive
spiking in the
ChR2 neurons.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
11
As currently implemented, the simultaneous application of VChR1 and ChR2
could be used to test progressive recruitment of different cell populations in
controlling
circuit behavior. For example, two different intemeuron or neuromodulatory
populations
could be recruited in stepwise fashion: first by isolating population A with
yellow light,
followed by driving the combination of populations A and B with added blue
light. This
kind of experiment has been the primary driving force behind developing
wavelength-
shifted channelrhodopsins, as the complexity of neural information processing
and
interactions of different neuromodulatory systems will require fast optical
excitation at
more than one wavelength to test the importance of combinatorial computations
and
modulatory gating in neural circuit dynamics and behavior.
While the role of single cell types can be tested in separate experiments, for

convenience in some experiments it might be useful to drive two isolated
populations at
different times. For this kind of experiment, an optimal strategy would entail
use of the
minimum 406 nm and 589 nm light intensities sufficient to separately trigger
reliable
ChR2 and VChR1 spikes respectively, which will simply require independent
calibration
in each experimental preparation (as in FIGs. 3A-C). A cross-taper of light
colors can
also be employed using a monochrometer or multiple filters; at yellow
wavelengths, the
VChR1 -labeled population will be exclusively controlled, and as the
excitation
wavelength becomes progressively more blue beyond 535 nm, the contribution of
the
ChR2-labeled population will become steadily more dominant (FIG. 1F).
Molecular
refinements (e.g., blueshifting ChR2 and narrowing the spectrum of VChR1) can
be
implemented to provide further separation at the blue end of the spectrum.
The identification and characterization of VChR1 for yellow-light neural
excitation here defines the third major functionally distinct category of fast
optogenetic
tools available for interrogating the organization and function of neural
circuits,
following the introduction of ChR2 for blue-light neural excitation and NpHR
for yellow-
light neural inhibition. In addition to its functionally significant red-
shifted action
spectrum, VChR1 displays additional properties that are of interest, including
reduced
ratio of peak to steady-state current (FIGs. 1C, 2B) compared with ChR2; while
typically
peak current magnitude in channelrhodopsins depends on light intensity,
external pH, and
membrane voltage, the steady-state to peak ratio is larger for VChR1 than ChR2
under all
conditions we have explored.

CA 02722278 2010-10-22
WO 2009/131837 12 PCT/US2009/039949
The existence of two independently controllable excitation proteins opens the
door
for a variety of applications including, but not limited to, applications for
treatment of a
variety of disorders and the use of a plurality of light-responsive proteins
that can be
selected so as to respond to a plurality of respective optical wavelengths.
The family of
single-component proteins has been shown to respond to multiple wavelengths
and
intensities of light. Aspects of the invention allow for further mutations
and/or searches
for sequences that allow for additional optical wavelengths and/or
individually
controllable protein channels. Variations on the optical stimulus (e.g., a
wavelength,
intensity or duration profile) can also be used. For instance, stimulation
profiles may
exploit overlaps in the excitation wavelengths of two different ion channel
proteins to
allow excitation of both proteins at the same time. In one such instance, the
proteins may
have different levels of responsibility. Thus, in a neural application, one
set of ion
channels may produce spiking at a different success percentage relative to a
second set of
ion channels.
Many human applications of the present invention require governmental approval
prior to their use. For instance, human use of gene therapy may require such
approval.
However, similar gene therapies in neurons (non-proliferative cells that are
non-
susceptible to neoplasms) are proceeding rapidly, with active, FDA-approved
clinical
trials already underway involving viral gene delivery to human brains. This is
likely to
facilitate the use of various embodiments of the present invention for a large
variety of
applications. The following is a non-exhaustive list of a few examples of such

applications and embodiments.
Addiction is associated with a variety of brain functions, including reward
and
expectation. Additionally, the driving cause of addiction may vary between
individuals.
According to one embodiment, addiction, for example nicotine addiction, may be
treated
with optogenetic stabilization of small areas on the insula. Optionally,
functional brain
imaging, for example cued-state PET or fMRI, may be used to locate a hyper
metabolic
focus in order to determine a precise target spot for the intervention on the
insula surface.
Optogenetic excitation of the nucleus accumbens and septum may provide reward
and pleasure to a patient without need for resorting to use of substances, and
hence may
hold a key to addiction treatment. Conversely, optogenetic stabilization of
the nucleus
accumbens and septum may be used to decrease drug craving in the context of
addiction.
In an alternative embodiment, optogenetic stabilization of hyper metabolic
activity

CA 02722278 2015-12-02
13
observed at the genu of the anterior cingulate (BA32) can be used to decrease
drug
craving. Optogenetic stabilization of cells within the arcuate nucleus of the
medial
hypothalamus which contain peptide products of pro-opiomelanocortin (POMC) and

cocaine-and-amphetamine-regulating transcript (CART) can also be used to
decrease
.. drug addiction behavior. For further information in this regard, reference
may be made to:
Naqvi NH, Rudrauf D, Darnasio H, Bechara A. "Damage to the insula disrupts
addiction
to cigarette smoking." Science. 2007 Jan 26; 315(5811):531-534.
Optogenetic stimulation of neuroendocrine neurons of the hypothalamic
.. periventricular nucleus that secrete somatostatin can be used to inhibit
secretion of growth
hormone from the anterior pituitary, for example in acromegaly. Optogenetic
stabilization of neuroendocrine neurons that secrete somatostatin or growth
hormone can
be used to increase growth and physical development. Among the changes that
accompany "normal" aging, is a sharp decline in serum growth hormone levels
after the
4th and 51h decades. Consequently, physical deterioration associated with
aging may be
lessened through optogenetic stabilization of the periventricular nucleus.
Optogenetic stabilization of the ventromedial nucleus of the hypothalamus,
particularly the pro-opiomelanocortin (POMC) and cocaine-and-amphetamine-
regulating
transcript (CART) of the arcuate nucleus, can be used to increase appetite,
and thereby
.. treat anorexia nervosa. Alternatively, optogenetic stimulation of the
lateral nuclei of the
hypothalamus can be used to increase appetite and eating behaviors.
Optogenetic excitation in the cholinergic cells of affected areas including
the
temporal lobe, the NBM (Nucleus basalis of Meynert) and the posterior
cingulate gyrus
(BA 31) provides stimulation, and hence neurotrophic drive to deteriorating
areas.
Because the affected areas are widespread within the brain, an analogous
treatment with
implanted electrodes may be less feasible than an opto-genetic approach.
Anxiety disorders are typically associated with increased activity in the left

temporal and frontal cortex and amygdal a, which trends toward normal as
anxiety
resolves. Accordingly, the affected left temporal and frontal regions and
amygdala may
be treated with optogenetic stabilization, so as to dampen activity in these
regions.
In normal physiology, photosensitive neural cells of the retina, which
depolarize
in response to the light that they receive, create a visual map of the
received light pattern.
Optogenetic ion channels can be used to mimic this process in many parts of
the body,

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
14
and the eyes are no exception. In the case of visual impairment or blindness
due to
damaged retina, a functionally new retina can be grown, which uses natural
ambient light
rather than flashing light patterns from an implanted device. The artificial
retina grown
may be placed in the location of the original retina (where it can take
advantage of the
optic nerve serving as a conduit back to the visual cortex). Alternatively,
the artificial
retina may be placed in another location, such as the forehead, provided that
a conduit for
the depolarization signals are transmitted to cortical tissue capable of
deciphering the
encoded information from the optogenetic sensor matrix. Cortical blindness
could also be
treated by simulating visual pathways downstream of the visual cortex. The
stimulation
would be based on visual data produced up stream of the visual cortex or by an
artificial
light sensor.
Treatment of tachycardia may be accomplished with optogenetic stimulation to
parasympathetic nervous system fibers including CN X or Vagus Nerve. This
causes a
decrease in the SA node rate, thereby decreasing the heart rate and force of
contraction.
Similarly, optogenetic stabilization of sympathetic nervous system fibers
within spinal
nerves Ti through T4, serves to slow the heart. For the treatment of
pathological
bradycardia, optogenetic stabilization of the Vagus nerve, or optogenetic
stimulation of
sympathetic fibers in Ti through T4 will serve to increase heart rate. Cardiac

disrhythmias resulting from aberrant electrical foci that outpace the
sinoatrial node may
be suppressed by treating the aberrant electrical focus with moderate
optogenetic
stabilization. This decreases the intrinsic rate of firing within the treated
tissue, and
permits the sinoatrial node to regain its role in pacing the heart's
electrical system. In a
similar way, any type of cardiac arrhythmia could be treated. Degeneration of
cardiac
tissue that occurs in cardiomyopathy or congestive heart failure could also be
treated
using this invention; the remaining tissue could be excited using various
embodiments of
the invention.
Optogenetic excitation stimulation of brain regions including the frontal
lobe,
parietal lobes and hippocampi, may increase processing speed, improve memory,
and
stimulate growth and interconnection of neurons, including spurring
development of
.. neural progenitor cells. As an example, one such application of the present
invention is
directed to optogenetic excitation stimulation of targeted neurons in the
thalamus for the
purpose of bringing a patient out of a near-vegetative (barely-conscious)
state. Growth of
light-gated ion channels or pumps in the membrane of targeted thalamus neurons
is

CA 02722278 2015-12-02
effected. These modified neurons are then stimulated (e.g., via optics which
may also
gain access by the same passageway) by directing a flash of light thereupon so
as to
modulate the function of the targeted neurons and/or surrounding cells. For
further
information regarding appropriate modulation techniques (via electrode-based
treatment)
5 or further information regarding the associated brain regions for such
patients, reference
may be made to: Schiff ND, Giacino JT, Kalmar K, Victor JD, Baker K, Gerber M,
Fritz
B, Eisenberg B, O'Connor JO, Kobylarz EJ, Farris S, Machado A, McCagg C, Plum
F,
Fins JJ, Rezai AR "Behavioral improvements with thalamic stimulation after
severe
traumatic brain injury," Nature, Vol. 448, Aug 2, 2007, pp. 600-604.
10 In an alternative embodiment, optogenetic excitation may be used to
treat
weakened cardiac muscle in conditions such as congestive heart failure.
Electrical
assistance to failing heart muscle of CHF is generally not practical, due to
the thin-
stretched, fragile state of the cardiac wall, and the difficulty in providing
an evenly
distributed electrical coupling between an electrodes and muscle. For this
reason,
15 preferred methods to date for increasing cardiac contractility have
involved either
pharmacological methods such as Beta agonists, and mechanical approaches such
as
ventricular assist devices. In this embodiment of the present invention,
optogenetic
excitation is delivered to weakened heart muscle via light emitting elements
on the inner
surface of a jacket surround the heart or otherwise against the affected heart
wall. Light
may be diffused by means well known in the art, to smoothly cover large areas
of muscle,
prompting contraction with each light pulse.
Optogenetic stabilization in the subgenual portion of the cingulate gyms
(Cg25),
yellow light may be applied with an implanted device. The goal would be to
treat
depression by suppressing target activity in manner analogous to what is
taught by
Mayberg HS et al., "Deep Brain Stimulation for Treatment-Resistant
Depression,"
Neuron, Vol. 45, 651-660, March 3, 2005, pp. 651-660.
In an alternative embodiment, an optogenetic excitation stimulation
method is to increase activity in that region in a manner analogous to what is
taught by
Schlaepfer et al., "Deep Brain stimulation to Reward Circuitry Alleviates
Anhedonia in
Refractory Major Depression," Neuropsychopharmaeology 2007, pp. 1-10.
In yet another embodiment, the left dorsolateral prefrontal cortex (LDPFC) is
targeted with an optogenetic excitation stimulation method. Pacing the LDLPFC
at 5-20

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
16
Hz serves to increase the basal metabolic level of this structure which, via
connecting
circuitry, serves to decrease activity in Cg 25, improving depression in the
process.
Suppression of the right dorsolateral prefrontal cortex (RDLPFC) is also an
effective
depression treatment strategy. This may be accomplished by optogenetic
stabilization on
the RDLPFC, or suppression may also be accomplished by using optogenetic
excitation
stimulation, and pulsing at a slow rate (e.g., 1 Hz or less) improving
depression in the
process. Vagus nerve stimulation (VNS) may be improved using an optogenetic
approach. Use of optogenetic excitation may be used in order to stimulate only
the vagus
afferents to the brain, such as the nodose ganglion and the jugular ganglion.
Efferents
.. from the brain would not receive stimulation by this approach, thus
eliminating some of
the side-effects of VNS including discomfort in the throat, a cough,
difficulty swallowing
and a hoarse voice. In an alternative embodiment, the hippocampus may be
optogenetically excited, leading to increased dendritic and axonal sprouting,
and overall
growth of the hippocampus. Other brain regions implicated in depression that
could be
treated using this invention include the amygdala, accumbens, orbitofrontal
and
orbitomedial cortex, hippocampus, olfactory cortex, and dopaminergic,
serotonergic, and
noradrenergic projections. Optogenetic approaches could be used to control
spread of
activity through structures like the hippocampus to control depressive
symptoms.
So long as there are viable alpha and beta cell populations in the pancreatic
islets
of Langerhans, the islets can be targeted for the treatment of diabetes. For
example, when
serum glucose is high (as determined manually or by closed loop glucose
detection
system), optogenetic excitation may be used to cause insulin release from the
beta cells of
the islets of Langerhans in the pancreas, while optogenetic stabilization is
used to prevent
glucagon release from the alpha cells of the islets of Langerhans in the
pancreas.
Conversely, when blood sugars are too low (as determined manually or by closed
loop
glucose detection system), optogenetic stabilization may be used to stop beta
cell
secretion of insulin, and optogenetic excitation may be used to increase alpha-
cell
secretion of glucagon.
For treatment of epilepsy, quenching or blocking epileptogenic activity is
.. amenable to optogenetic approaches. Most epilepsy patients have a
stereotyped pattern of
activity spread resulting from an epileptogenic focus. Optogenetic
stabilization could be
used to suppress the abnormal activity before it spreads or truncated it early
in its course.
Alternatively, activation of excitatory tissue via optogenetic excitation
stimulation could

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
17
be delivered in a series of deliberately asynchronous patterns to disrupt the
emerging
seizure activity. Another alternative involves the activation of optogenetic
excitation
stimulation in GABAergic neurons to provide a similar result. Thalamic relays
may be
targeted with optogenetic stabilization triggered when an abnoinial EEG
pattern is
detected.
Another embodiment involves the treatment of gastrointestinal disorders. The
digestive system has its own, semi-autonomous nervous system containing
sensory
neurons, motor neurons and interneurons. These neurons control movement of the
GI
tract, as well as trigger specific cells in the gut to release acid, digestive
enzymes, and
hormones including gastrin, cholecystokinin and secretin. Syndromes that
include
inadequate secretion of any of these cellular products may be treated with
optogenetic
stimulation of the producing cell types, or neurons that prompt their
activity. Conversely,
optogenetic stabilization may be used to treat syndromes in which excessive
endocrine
and exocrine products are being created. Disorders of lowered intestinal
motility, ranging
from constipation (particularly in patients with spinal cord injury) to
megacolan may be
treated with optogenetic excitation of motor neurons in the intestines.
Disorders of
intestinal hypermotility, including some forms of irritable bowel syndrome may
be treated
with optogenetic stabilization of neurons that control motility. Neurogentic
gastric outlet
obstructions may be treated with optogenetic stabilization of neurons and
musculature in
the pyloris. An alternative approach to hypomobility syndromes would be to
provide
optogenetic excitation to stretch-sensitive neurons in the walls of the gut,
increasing the
signal that the gut is full and in need of emptying.
In this same paradigm, an approach to hypermobility syndromes of the gut would

be to provide optogenetic stabilization to stretch receptor neurons in the
lower GI, thus
providing a "false cue" that the gut was empty, and not in need of emptying.
In the case
of frank fecal incontinence, gaining improved control of the internal and
external
sphincters may be preferred to slowing the motility of the entire tract.
During periods of
time during which a patient needs to hold feces in, optogenetic excitation of
the internal
anal sphincter will provide for retention. Providing optogenetic stimulation
to the
external sphincter may be used to provide additional continence. When the
patient is
required to defecate, the internal anal sphincter, and then external anal
sphincter should
be relaxed, either by pausing the optogenetic stimulation, or by adding
optogenetic
stabilization.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
18
Conductive hearing loss may be treated by the use of optical cochlear
implants.
Once the cochlea has been prepared for optogenetic stimulation, a cochlear
implant that
flashes light may be used. Sensorineural hearing loss may be treated through
optical
stimulation of downstream targets in the auditory pathway.
Another embodiment of the present invention is directed toward the treatment
of
blood pressure disorders, such as hypertension. Baroreceptors and
chemoreceptors in
regions such as the aorta (aortic bodies and paraaortic bodies) and the
carotid arteries
("carotic bodies") participate in the regulation of blood pressure and
respiration by
sending afferents via the vagus nerve (CN X), and other pathways to the
medulla and
pons, particularly the solitary tract and nucleus. Optogenetic excitation of
the carotid
bodies, aortic bodies, paraortic bodies, may be used to send a false message
of
"hypertension" to the solitary nucleus and tract, causing it to report that
blood pressure
should be decreased. Optogenetic excitation or stabilization directly to
appropriate parts
of the brainstem may also be used to lower blood pressure. The opposite
modality causes
the optogenetic approach to serve as a pressor, raising blood pressure. A
similar effect
may also be achieved via optogenetic excitation of the Vagus nerve, or by
optogenetic
stabilization of sympathetic fibers within spinal nerves T1 -T4. In an
alternative
embodiment, hypertension may be treated with optogenetic stabilization of the
heart,
resulting in decreased cardiac output and lowered blood pressure. According to
another
.. embodiment, optogenetic stabilization of aldosterone-producing cells within
the adrenal
cortex may be used to decrease blood pressure. In yet another alternative
embodiment,
hypertension may be treated by optogenetic stabilization of vascular smooth
muscle.
Activating light may be passed transcutaneously to the peripheral vascular
bed.
Another example embodiment is directed toward the treatment of hypothalamic-
pituitary-adrenal axis disorders. In the treatment of hypothyroidism,
optogenetic
excitation of parvocellular neuroendocrine, neurons in the paraventricular and
anterior
hypothalamic nuclei can be used to increase secretion of thyrotropin-releasing
hormone
(TRH). TRH, in turn, stimulates anterior pituitary to secrete TSH. Conversely,

hyperthyroidism may be treated with optogenetic stabilization of the
provocellular
neuroendocrine neurons. For the treatment of adrenal insufficiency, or of
Addison's
disease, optogenetic excitation of parvocellular neuroendocrine neurons in the
supraoptic
nucleus and paraventricular nuclei may be used to increase the secretion of
vasopressin,
which, with the help of corticotropin-releasing hormone (CRH), stimulate
anterior

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
19
pituitary to secrete ACTH. Cushing syndrome, frequently caused by excessive
ACTH
secretion, may be treated with optogenetic stabilization of the parvocellular
neuroendocrine neurons of supraoptic nucleus via the same physiological chain
of effects
described above. Neuroendocrine neurons of the arcuate nucleus produce
dopamine,
which inhibits secretion of prolactin from the anterior pituitary.
Hyperprolactinemia can
therefore be treated via optogenetic excitation, while hypoprolactinemia can
be treated
with optogenetic stabilization of the neuroendocrine cells of the arcuate
nucleus.
In the treatment of hyperautonomic states, for example anxiety disorders,
optogenetic stabilization of the adrenal medulla may be used to reduce
norepinephrine
output. Similarly, optogenetic stimulation of the adrenal medulla may be used
in persons
with need for adrenaline surges, for example those with severe asthma, or
disorders that
manifest as chronic sleepiness.
Optogenetic stimulation of the adrenal cortex will cause release of chemicals
including cortisol, testosterone, and aldosterone. Unlike the adrenal
medualla, the adrenal
cortex receives its instructions from neuroendocrine hormones secreted from
the pituitary
and hypothalamus, the lungs, and the kidneys. Regardless, the adrenal cortex
is amenable
to optogenetic stimulation. Optogenetic stimulation of the cortisol-producing
cells of the
adrenal cortex may be used to treat Addison's disease. Optogenetic
stabilization of
cortisol-producing cells of the adrenal cortex may be used to treat Cushing's
disease.
Optogenetic stimulation of testosterone-producing cells may be used to treat
disorders of
sexual interest in women: Optogenetic stabilization of those same cells may be
used to
decrease facial hair in women. Optogenetic stabilization of aldosterone-
producing cells
within the adrenal cortex may be used to decrease blood pressure. Optogenetic
excitation
of aldosterone-producing cells within the adrenal cortex may be used to
increase blood
pressure.
Optogenetic excitation stimulation of specific affected brain regions may be
used
to increase processing speed, and stimulate growth and interconnection of
neurons,
including spurring the maturation of neural progenitor cells. Such uses can be
particularly useful for treatment of mental retardation.
According to another embodiment of the present invention, various muscle
diseases and injuries can be treated. Palsies related to muscle damage,
peripheral nerve
damage and to dystrophic diseases can be treated with optogenetic excitation
to cause
contraction, and optogenetic stabilization to cause relaxation. This latter
relaxation via

CA 02722278 2010-10-22
WO 2009/131837
PCT/US2009/039949
optogenetic stabilization approach can also be used to prevent muscle wasting,
maintain
tone, and permit coordinated movement as opposing muscle groups are
contracted.
Likewise, frank spasticity can be treated via optogenetic stabilization.
In areas as diverse as peripheral nerve truncation, stroke, traumatic brain
injury
5 and spinal cord injury, there is a need to foster the growth of new
neurons, and assist with
their integration into a functional network with other neurons and with their
target tissue.
Re-growth of new neuronal tracts may be encouraged via optogenetic excitation,
which
serves to signal stem cells to sprout axons and dendrites, and to integrate
themselves with
the network. Use of an optogenetic technique (as opposed to electrodes)
prevents receipt
10 of signals by intact tissue, and serves to ensure that new target tissue
grows by virtue of a
communication set up with the developing neurons, and not with an artificial
signal like
current emanating from an electrode.
Obesity can be treated with optogenetic excitation to the ventromedial nucleus
of
the hypothalamus, particularly the pro-opiomelanocortin (POMC) and cocaine-and-

15 amphetamine-regulating transcript (CART) of the arcuate nucleus. In an
alternative
embodiment, obesity can be treated via optogenetic stabilization of the
lateral nuclei of
the hypothalamus. In another embodiment, optogenetic stimulation to leptin-
producing
cells or to cells with leptin receptors within the hypothalamus may be used to
decrease
appetite and hence treat obesity.
20
Destructive lesions to the anterior capsule and analogous DBS to that region
are
established means of treating severe, intractable obsessive-compulsive
disorder 48
(0CD48). Such approaches may be emulated using optogenetic stabilization to
the
anterior limb of the internal capsule, or to regions such as BA32 and Cg24
which show
metabolic decrease as OCD remits.
Chronic pain can be treated using another embodiment of the present invention.
Electrical stimulation methods include local peripheral nerve stimulation,
local cranial
nerve stimulation and "sub threshold" motor cortex stimulation. Reasonable
autogenic
approaches include optogenetic stabilization at local painful sites. Attention
to promoter
selection would ensure that other sensory and motor fibers would be
unaffected.
Selective optogenetic excitation of interneurons at the primary motor cortex
also may
provide effective pain relief. Also, optogenetic stabilization at the sensory
thalamus,
(particularly medial thalamic nuclei), periventricular grey matter, and
ventral raphe nuclei,
may be used to produce pain relief. In an alternative embodiment, optogenetic

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
21
stabilization of parvalbumin-expressing cells targeting as targeting strategy,
may be used
to treat pain by decreasing Substance P production. The release of endogenous
opiods
may be accomplished by using optogenetic excitation to increase activity in
the nucleus
accumbens. In an alternative embodiment, when POMC neurons of the arcuate
nucleus
of the medial hypothalamus are optogenetically excited, beta endorphin are
increased,
providing viable treatment approaches for depression and for chronic pain.
Certain personality disorders, including the borderline and antisocial types,
demonstrate focal deficits in brain disorders including "hypofrontality."
Direct or indirect
optogenetic excitation of these regions is anticipated to produce improvement
of
symptoms. Abnormal bursts of activity in the amygdala are also known to
precipitate
sudden, unprompted flights into rage: a symptom of borderline personality
disorder, as
well as other conditions, which can benefit from optogenetic stabilization of
the amygdala.
Optogenetic approaches could improve communication and synchronization between

different parts of the brain, including amygdala, striatum, and frontal
cortex, which could
help in reducing impulsiveness and improving insight.
The amygdalocentric model of post-traumatic-stress disorder (PTSD) proposes
that it is associated with hyperarousal of the amygdala and insufficient top-
down control
by the medial prefrontal cortex and the hippocampus. Accordingly, PTSD may be
treated with optogenetic stabilization of the amygdale or hippocampus.
Schizophrenia is characterized by abnormalities including auditory
hallucinations.
These might be treated by suppression of the auditory cortex using optogenetic

stabilization. Hypofrontality associated with schizophrenia might be treated
with
optogenetic excitation in the affected frontal regions. Optogenetic approaches
could
improve communication and synchronization between different parts of the brain
which
could help in reducing misattribution of self-generated stimuli as foreign.
Optogenetic stabilization of cells within the arcuate nucleus of the medial
hypothalamus, which contain peptide products of pro-opiomelanocortin (POMC)
and
cocaine-and-amphetamine-regulating transcript (CART), can be used to reduce
compulsive sexual behavior. Optogenetic excitation of cells within the arcuate
nucleus of
the medial hypothalamus which contain peptide products of pro-opiomelanocortin
(POMC) and cocaine-and-amphetamine-regulating transcript (CART) may be used to

increase sexual interest in the treatment of cases of disorders of sexual
desire. In the
treatment of disorders of hypoactive sexual desire testosterone production by
the testes

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
22
and the adrenal glands can be increased through optogenetic excitation of the
pituitary
gland. Optogenetic excitation of the nucleus accumbens can be used for the
treatment of
anorgasmia.
The suprachiasmatic nucleus secretes melatonin, which serves to regulate
sleep/wake cycles. Optogenetic excitation to the suprachiasmic nucleus can be
used to
increase melatonin production, inducing sleep, and thereby treating insomnia.
Orexin
(hypocretin) neurons strongly excite numerous brain nuclei in order to promote

wakefulness. Optogentetic excitation of orexin-producing cell populations can
be used to
treat narcolepsy, and chronic daytime sleepiness.
Optogenetic stimulation of the supraoptic nucleus may be used to induce
secretion
of oxytocin, can be used to promote parturition during childbirth, and can be
used to treat
disorders of social attachment.
Like muscular palsies, the motor functions that have been de-afferented by a
spinal cord injury may be treated with optogenetic excitation to cause
contraction, and
optogenetic stabilization to cause relaxation. This latter relaxation via
optogenetic
stabilization approach may also be used to prevent muscle wasting, maintain
tone, and
permit coordinated movement as opposing muscle groups are contracted.
Likewise, frank
spasticity may be treated via optogenetic stabilization. Re-growth of new
spinal neuronal
tracts may be encouraged via optogenetic excitation, which serves to signal
stem cells to
sprout axons and dendrites, and to integrate themselves with the network.
Stroke deficits include personality change, motor deficits, sensory deficits,
cognitive loss, and emotional instability. One strategy for the treatment of
stroke deficits
is to provide optogenetic stimulation to brain and body structures that have
been
deafferented from excitatory connections. Similarly, optogenetic stabilization
capabilities
can be imparted on brain and body structures that have been deafferented from
inhibitory
connections.
Research indicates that the underlying pathobiology in Tourette's syndrome is
a
phasic dysfunction of dopamine transmission in cortical and subcortical
regions, the
thalamus, basal ganglia and frontal cortex. In order to provide therapy,
affected areas are
preferably first identified using techniques including functional brain
imaging and
magnetoencephalography (MEG). Whether specifically identified or not,
optogenetic
stabilization of candidate tracts may be used to suppress motor tics. Post-
implantation

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
23
empirical testing of device parameters reveals which sites of optogenetic
stabilization,
and which are unnecessary to continue.
In order to treat disorders of urinary or fecal incontinence optogenetic
stabilization
can be used to the sphincters, for example via optogenetic stabilization of
the bladder
detrussor smooth muscle or innervations of that muscle. When micturation is
necessary,
these optogenetic processes are turned off, or alternatively can be reversed,
with
optogenetic stabilization to the (external) urinary sphincter, and optogenetic
excitation of
the bladder detrussor muscle or its innervations. When a bladder has been
deafferentated,
for example, when the sacral dorsal roots are cut or destroyed by diseases of
the dorsal
roots such as tabes dorsalis in humans, all reflex contractions of the bladder
are abolished,
and the bladder becomes distended. Optogenetic excitation of the muscle
directly can be
used to restore tone to the detrussor, prevent kidney damage, and to assist
with the
micturition process. As the bladder becomes "decentralized" and hypersensitive
to
movement, and hence prone to incontinence, optogenetic stabilization to the
bladder
muscle can be used to minimize this reactivity of the organ.
In order to selectively excite/inhibit a given population of neurons, for
example
those involved in the disease state of an illness, several strategies can be
used to target the
optogenetic proteins/molecules to specific populations.
For various embodiments of the present invention, genetic targeting may be
used
to express various optogenetic proteins or molecules. Such targeting involves
the
targeted expression of the optogenetic proteins/molecules via genetic control
elements
such as promoters (e.g., Parvalbumin, Somatostatin, Cholecystokinin, GFAP),
enhancers/silencers (e.g., Cytomaglovirus Immediate Early Enhancer), and other

transcriptional or translational regulatory elements (e.g., Woodchuck
Hepatitis Virus
Post-transcriptional Regulatory Element). Permutations of the
promoter+enhancer+regulatory element combination can be used to restrict the
expression of optogenetic probes to genetically-defined populations.
Various embodiments of the present invention may be implemented using
spatial/anatomical targeting. Such targeting takes advantage of the fact that
projection
patterns of neurons, virus or other reagents carrying genetic information (DNA
plasmids,
fragments, etc), can be focally delivered to an area where a given population
of neurons
project to. The genetic material will then be transported back to the bodies
of the neurons
to mediate expression of the optogenetic probes. Alternatively, if it is
desired to label

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
24
cells in a focal region, viruses or genetic material may be focally delivered
to the
interested region to mediate localized expression.
Various gene delivery systems are useful in implementing one or more
embodiments of the present invention. One such delivery system is Adeno-
Associated
Virus (AAV). AAV can be used to deliver a promoter+optogenetic probe cassette
to a
specific region of interest. The choice of promoter will drive expression in a
specific
population of neurons. For example, using the CaMKIIa promoter will drive
excitatory
neuron specific expression of optogenetic probes. AAV will mediate long-term
expression of the optogenetic probe for at least one year or more. To achieve
more
specificity, AAV may be pseudotyped with specific serotypes 1 to 8, with each
having
different trophism for different cell types. For instance, serotype 2 and 5 is
known to
have good neuron-specific trophism.
Another gene delivery mechanism is the use of a retrovirus. HIV or other
lentivirus-based retroviral vectors may be used to deliver a
promoter+optogenetic probe
cassette to a specific region of interest. Retroviruses may also be pseudo-
typed with the
Rabies virus envelope glycoprotein to achieve retrograde transport for
labeling cells
based on their axonal projection patterns. Retroviruses integrate into the
host cell's
genome, therefore are capable of mediating permanent expression of the
optogenetic
probes. Non-lentivirus based retroviral vectors can be used to selectively
label dividing
cells.
Gutless Adenovirus and Herpes Simplex Virus (HSV) are two DNA-based viruses
that can be used to deliver promoter+optogenetic probe cassette into specific
regions of
the brain as well. HSV and Adenovirus have much larger packaging capacities
and
therefore can accommodate much larger promoter elements and can also be used
to
deliver multiple optogenetic probes or other therapeutic genes along with
optogenetic
probes.
Focal Electroporation can also be used to transiently transfect neurons. DNA
plasmids or fragments can be focally delivered into a specific region of the
brain. By
applying mild electrical current, surrounding local cells will receive the DNA
material
and expression of the optogenetic probes.
In another instance, lipofection can be used by mixing genetic material with
lipid
reagents and then subsequently injected into the brain to mediate transfection
of the local
cells.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
Various embodiments involve the use of various control elements. In addition
to
genetic control elements, other control elements (particularly promoters and
enhancers
whose activities are sensitive to chemical, magnetic stimulation or infrared
radiation) can
be used to mediate temporally-controlled expression of the optogenetic probes.
For
5 example, a promoter whose transcriptional activity is subject to infrared
radiation allows
one to use focused radiation to fine tune the expression of optogenetic probes
in a focal
region at only the desired time.
Parkinson's Disease can be treated by expressing optogenetic stabilization in
the
glutamatergic neurons in either the subthalamic nucleus (STN) or the globus
pallidus
10 interna (GPi) using an excitatory-specific promoter such as CaMKIIa, and
apply
optogenetic stabilization. Unlike electrical modulation in which all cell-
types are affected,
only glutamatergic STN neurons would be suppressed.
Aspects of the present invention are directed towards testing a model of a
neural
circuit or disease. The model can define output response of the circuit as a
function of
15 input signals. The output response can be assessed using a number of
different
measurable characteristics. For instance, characteristics can include an
electrical response
of downstream neurons and/or behavioral response of a patient. To test the
model,
optogentic probes are expressed at an input position for the model. The
optogenetic
probes are stimulated and the output characteristics are monitored and
compared to an
20 output predicted by the model.
In certain implementations, the use of optogenetic probes allows for fine
tuning of
models defined using electrical probes. As electrical probes provide only
limited ability
to direct the stimulus and thus are not well suited for stimulus of certain
areas without
also directly stimulating nearby areas. Optogenetic probes disclosed herein
provide a
25 mechanism for more precise selection of the stimulus location. For
instance, the stimulus
from the optogenetic probes can be directed to very specific types of
circuits/cells, such
as afferent fibers. The following description provides an example
implementation
consistent with such an embodiment and is meant to show the feasibility and
wide-
ranging applicability for aspects of present invention.
According to one embodiment of the present invention, the invention may be
used
in animal models of DBS, for example in Parkinsonian rats, to identify the
target cell
types responsible for therapeutic effects (an area of intense debate and
immense clinical

CA 02722278 2015-12-02
26
importance). This knowledge alone may lead to the development of improved
pharmacological and surgical strategies for treating human disease.
One such application involves long-term potentiation (LIP) and/or long-term
depression (LTD) between two neural groups. By targeting the expression of
VChR1 and
ChR2 to different neural populations and stimulating each with a different
frequency of
LTP or LTD can be accomplished between the two groups. Each group can be
individually controlled using the respective wavelength of light. This can be
particularly
useful for applications in which the spatial arrangement of the two groups
presents issues
with individual control using the same wavelength of light. Thus, the light
delivery
device(s) are less susceptible to exciting the wrong neural group and can be
less reliant
upon precise spatial location of the optical stimulus.
The delivery of the proteins to cells in vivo can be accomplished using a
number
of different deliver devices, methods and systems. On such delivery device is
an
implantable device that delivers a nucleotide sequence for modifying cells in
vivo, such as
a viral-vector. The implantable device can also include a light delivery
mechanism. The
light delivery can be accomplished using, for example, light-emitting diodes
(LEDs),
fiber optics and/or Lasers.
Another embodiment of the present invention involves the use of VCIaR1 in
affecting stem cell fate including survival/death, differentiation and
replication. The
modulation of electrical properties has been shown to control stem cell fate.
Various
techniques can be used to provide stimulus patterns that modify stem cell
fate. A specific
example is consistent with those techniques used in Deisseroth, K. et al.
"Excitation-
neurogenesis coupling in adult neural stein/progenitor cells," Neuron 42, pp.
535-552
(2004).
Another embodiment of the present invention is directed to the use of VChR1 to

assess the efficacy of treatments. This can include, but is not limited to,
drug screening,
treatment regimens or modeling of treatments/disorders. In a specific
embodiment,
VChR1 is used as the primary optically responsive protein in such assessments.
In
alternate embodiments, VChR1 is used with other types of optically responsive
proteins
(e.g., ChR2 and/or NpHR) that respond to different wavelengths.
A specific embodiment of the present invention involves the use of VChR1 to
generate a mammalian codon-optimized cDNA sequence and synthesized (DNA 2.0,
Menlo Park, CA).
=
=

CA 02722278 2015-12-02
27
Lentiviral vector construction was accomplished using the following methods.
VChRl-EYFP was constructed by fusing VChR1(1-300) with EYFP via a NotI
restriction
site. The fusion gene was then ligated into the AgeI and EcoRI sites of alpha-
CaMKII
lentiviral backbone to generate the pLenti-CaMKIIa-VChRl-EYFP-WPRE vector.
Construction of the pLenti-CaMKIIa-ChR2-EYFP-WPRE vector was previously
described. Recombinant lentiviruses were generated. For further details
regarding the
construction or use of such vectors reference can be made to Zhang, F., et al.

"Multimodal fast optical interrogation of neural circuitry," Nature 446, pp.
633-639
(2007).
Cultured hippocampal neurons were prepared as described in Boyden, E.S.,
Zhang, F., Bamberg, E., Nagel, G. & Deisseroth, K. "Millisecond-timescale,
genetically
targeted optical control of neural activity," Nat Neurosci 8, pp. 1263-1268
(2005).
For whole-cell recording in cultured hippocampal neurons, the intracellular
solution contained 129 mM K-Gluconate, 10 mM HEPES, 10 mM KC1, 4 mM MgATP,
and 0.3 mM Na3GTP, titrated to pH 7.2. For cultured hippocampal neurons,
Tyrode's
solution Was employed as the extracellular solution (125 mM NaCl, 2 mM KC1, 3
mM
CaCl2, 1 mM MgC12, 30 mM glucose, and 25 mM BEPES, titrated to pH 7.3).
Recordings were conducted on an upright Leica DM-LFSA microscope equipped with
a
40X water-immersion objective. Borosilicate glass (Sutter Instruments) pipette
resistances were ¨5 Ma, with a range of 4-6 M. Access resistance was 10-30 ML
and
monitored for stability throughout the recording. All recordings were
conducted in the
presence of synaptic transmission blockers as described in Boyden, E.S.,
Zhang, F.,
Bamberg, E., Nagel, G. 84 Deisseroth, K. "Millisecond-timescale, genetically
targeted
optical control of neural activity," Nat Neurosci 8, pp. 1263-1268 (2005).
For hippocampal neuron photostimulation, the following three filters were used
in
the Lambda DG-4 optical switch (Sutter Instruments) with a 300W Xenon lamp:
406nm
(FF01-406/15-25), 531m (FF01-531/22-25), and 589nm (FF01-589/15-25) (Semrock).
For oocyte experiments, a synthetic DNA sequence corresponding to VC111211-
313 (vehopl ; adapted to human codon-usage, Geneart, Regensburg, Germany) was
subcloned into VChR1 pGEMHE and pEGFP. cRNAs encoding ChR2 and VChRl,
synthesized in vitro from pGEMHEplasmid by T7 RNA polymerase (mMessage
mMachine, Ambion), were injected into the oocytes (50 ng/cell). The oocytes
were

CA 02722278 2010-10-22
WO 2009/131837
PCT/US2009/039949
28
stored for 3-7 days in the dark at 18 C in Ringer solution (96 mM NaC1, 5 mM
KC1,
1.8 mM CaCl2, 1 mM MgCl2, 5 mM MOPS-NaOH, pH 7.5) in the presence of 1 mg/ml
penicillin, 1 mg/ml streptomycin, 1 M all-trans-retinal, and 0.5 mM
theophylline.
Two-electrode voltage clamp on Xenopus laevis oocytes were performed to obtain
action spectra. 10 ns laser flashes (400-620 nm, 4-9 x 1019 photons s-1 m-2)
from a
Rainbow OPO (OPOTEK, Carlsbad, CA) pumped by the third harmonic of a Brilliant
b
Nd-YAG-Laser (Quantel, Les Ulis Cedex, France) were applied to the oocyte via
a 1 mm
light guide. The amplifier Tec-05X (NPI Electronic, Tamm, Germany) was
compensated
to keep the voltage change below 0.05 mV at a half saturating laser flash.
Data
acquisition and light triggering were controlled with pCLAMP software via
DigiData
1440A interfaces (Molecular Devices, Sunnyvale, USA).
The following discussion includes a detailed discussion regarding results of
an
application for treatment and characterization of Parkinson's Disease (PD).
This specific
implementation and the corresponding results are not meant to be limiting.
To first address the most widely-held hypothesis in the field, we asked if
direct,
reversible, bona fide inhibition of local-circuit excitatory STN neurons would
be
therapeutic in PD. The STN measures <1mm3 in rats, but targeting accuracy can
be aided
by extracellular recordings during opsin vector introduction, since STN is
characterized
by a particular firing pattern which is distinguishable from bordering regions
(FIG. 4A,
FIG. 10C).
The STN is a predominantly excitatory structure embedded within an inhibitory
network. This anatomical arrangement enables a targeting strategy for
selective STN
inhibition (FIG. 4B), in which eNpHR is expressed under control of the
CaMKIIa promoter, selective for excitatory glutamatergic neurons and not
inhibitory
cells, fibers of passage, glia, or neighboring structures. In this way true
optical inhibition
is targeted to the dominant local neuron type within STN.
Optical circuit interventions were tested in rats that had been made
hemiparkinsonian by injection of 6-hydroxydoparnine (6-0HDA) unilaterally into
the
right medial forebrain bundle (MFB). Loss of nigral dopaminergic cells
following 6-
OHDA administration was confirmed by decreased tyrosine hydroxylase levels
unilaterally in the substantia nigra pars compacta (FIG. 10A). These
hemiparkinsonian
rodents have specific deficits in contralateral (left) limb use and display
(rightward)
rotations ipsilateral to the lesion, which increase in frequency when the
subjects are given

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
29
amphetamine to facilitate functional evaluation, and decrease in frequency
upon treatment
with dopamine agonists or following electrical DBS (FIG. 4D, right). This
amphetamine-
induced rotation test is widely used for identifying treatments in
hemiparkinsonian
rodents, which can be complemented with other behavioral assays such as
locomotion,
climbing, and head position bias. To directly inhibit the excitatory STN
neurons, we
delivered lentiviruses carrying eNpHR under the CaMKIIa promoter to the right
STN of
the hemiparkinsonian rats. CaMKIIa::eNpHR-EYFP expression was specific to
excitatory
neurons (as shown by CaMKIIa and glutamate expression; FIG. 4B, right; FIG.
11A),
robust (95.73% 1.96 s.e.m infection rate assessed in n = 220 CaMKIIa
positive cells),
and restricted to the STN (FIG. 4B, left and middle). To validate the
resulting
physiological effects of optical control, a hybrid optical
stimulation/electrical recording
device (optrode) was employed in isoflurane-anesthetized animals to confirm
that eNpHR
was functional in vivo, potently inhibiting (>80%) spiking of recorded neurons
in the STN
(FIG. 4C; FIG. S13A, B; FIG. 14A). This cell type-targeted inhibition was
temporally
precise and reversible, and extended across all frequency bands of neuronal
firing
(FIG. 4C, FIG. 16A).
For behavioral rotation assays in the hemiparkinsonian rats, the STN-targeted
fiberoptic was coupled to a 561nm laser diode to drive eNpHR. Electrical DBS
was
highly effective at reducing pathological rotational behavior, but despite
precise targeting
and robust physiological efficacy of eNpHR inhibition, the hemiparkinsonian
animals did
not show even minimal changes in rotational behavior with direct true optical
inhibition
of the local excitatory STN neurons (FIG. 4D). In addition, there was no
effect on path
length and head position bias in response to light during these experiments
(see
supplementary methods). While muscimol and lidocaine administration to the
region of
the STN in monkeys and rodents can relieve Parkinsonian symptoms (30), the
data in
FIG. 4 show that the more specific intervention of selectively decreasing
activity in
excitatory local neurons of the STN appeared not sufficient by itself to
affect motor
symptoms.
Another possibility is that DBS could be driving secretion of glial modulators
which would have the capability to modulate local STN circuitry; this would be
consistent with recent findings indicating that a glial-derived factor
(adenosine)
accumulates during DBS and plays a role in DBS-mediated attenuation of
thalamic
tremor. Indeed, the STN expresses receptors for glia-derived modulators which
can

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
inhibit postsynaptic currents in the STN. ChR2 presents an interesting
possibility for
recruitment of glia; when opened by light, in addition to Na+ and K ions,
ChR2 can also
pass trace Ca2+ currents that trigger Ca2+ waves in and activate ChR2-
expressing
astroglia. We employed a GFAP promoter to target ChR2 to local astroglia,
validated
5 with GFAP and S10013 staining (FIG. 5A, FIG. 11B). Optrode recordings
revealed that
blue light stimulation of STN following transduction with GFAP::ChR2
reversibly
inhibited neuronal firing in the STN (FIG. 5B, FIG. 12A), with variable delay
on the
timescale of seconds. However, recruiting astroglial cells by this mechanism
was not
sufficient to cause even trace responses in motor pathology in parkinsonian
rodents (FIG.
10 5C, FIG. 12B). Path length and head position bias were also not affected
by light during
these experiments. While these data do not exclude the importance of local STN

inhibition as a contributing factor in DBS response, as not all STN neurons
may be
affected in the same way by indirect glial modulation, the direct activation
of local glial
cells appeared not sufficient to treat parkinsonian symptoms, pointing to
other circuit
15 mechanisms.
Network oscillations at particular frequencies could play important roles in
both
PD pathology and treatment. For example, PD is characterized by pathological
levels of
beta oscillations in the basal ganglia, and synchronizing STN at gamma
frequencies may
ameliorate PD symptoms while beta frequencies may worsen symptoms. Because
simple
20 inhibition of excitatory cell bodies in the STN did not affect
behavioral pathology, and
since high-frequency stimulation (HFS: 90-130 Hz) is used for electrical DBS,
we used
ChR2 to drive high-frequency oscillations in this range within the STN. We
injected
CaMKIIa::ChR2 into the STN (FIG. 6A) and used pulsed illumination with a 473nm

laser diode to activate excitatory neurons in the STN (FIG. 6B, FIG. 14B)
during
25 behavioral testing in parkinsonian rodents (FIG. 6C, FIG. 12C). Despite
robust effects on
high-frequency power of neuronal spike rate in STN of anesthetized animals
(FIG. 16B),
HFS delivered locally to the STN area failed to affect PD behavioral symptoms
(path
length and head position bias were unchanged by light - see supplementary
methods).
Animals tested in parallel with beta frequency pulses also showed no
behavioral response,
30 indicating that (while not excluding a contributory role) directly
generated oscillations
within the STN excitatory neurons are not sufficient to account for
therapeutic effects.
We have previously measured in cortical and hypothalamic tissue the
propagation
of blue light in the setting of laser diode-fiberoptic illumination; we
observed that

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
31
substantial tissue volumes (comparable to that of the STN) could reliably be
recruited at a
light power density sufficient to drive physiologically significant microbial
opsin
currents. It was important to repeat and extend these measurements to the PD
setting.
First, we confirmed that the propagation measurements of blue light (473 nm)
in brain
tissue represent a lower bound on the volume of tissue recruited, due to
reduced scattering
of lower-energy photons delivered from the 561m laser diode; therefore
sufficient light
power is present to activate opsins within 1.5mm of the fiber for either
wavelength of
light (FIG. 7A). We next extended these findings with a functional assay for
tissue
recruitment under conditions mimicking our behavioral experiments (FIG. 7B,C).
After
an in vivo optical stimulation paradigm targeted to the CaMKIIa::ChR2
expressing STN
in freely moving rats, we performed immunohistochemistry for c-fos, a
biochemical
marker of neuronal activation. We observed robust c-fos activation in STN
(FIG. 7B)
over a widespread volume (FIG. 7C); indeed, as expected from our light
scattering
measurements and tissue geometry, we found that at least 0.7 mm3 of STN is
recruited by
light stimulation, closely matching the actual volume of the STN (FIG. 7C).
Therefore,
light penetration was not limiting since the entire STN is recruited by the
optical
modulation paradigms of Figs. 4-6.
Therapeutic effects could arise from driving axonal projections that enter the
STN,
as DBS electrodes will potently modulate not just local cells and their
efferents, but also
afferent fibers. Optogenetics discriminates these two possibilities, as the
lentiviruses
transduce somata without transducing afferent axons. To assess the possibility
that PD
motor behavioral responses are modulated by targeting afferent projections to
the STN,
we used Thy 1 ::ChR2 transgenic mice in which ChR2 is expressed in projection
neurons,
and we verified that in Thyl ::ChR2 line 18, ChR2-YFP is excluded from cell
bodies in
the STN but is abundant in afferent fibers (FIG. 8A).
We conducted optrode recordings in anesthetized 6-0HDA mice (FIG. 10B) to
assess local effects on STN physiology of driving afferent axons selectively,
and found
frequency-dependent effects (FIG. 8B). First, we observed that HFS of afferent
fibers to
the STN potently reduced STN spiking across all frequency bands; this effect
did not
completely shut down local circuitry, as low-amplitude high-frequency
oscillations
persisted during stimulation (FIG. 8B; FIG. 13C, D; FIG. 14C). Next, we found
that LFS
of afferent fibers increased beta-frequency firing in the STN without
affecting
endogenous bursting (FIG. 8B, FIG. 14D). We next assessed the impact of these
specific

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
32
interventions on PD behavior in 6-0HDA mice, and for the first time among the
optogenetic interventions, we observed marked effects. Driving STN afferent
fibers with
HFS robustly and reversibly ameliorated PD symptoms, measured by rotational
behavior
and head position bias (FIG. 8C). The HFS effects were not subtle; indeed, in
nearly
every case these severely parkinsonian animals were restored to behavior
indistinguishable from normal, and in every case the therapeutic effect
immediately and
fully reversed, with return of ipsilateral rotations upon discontinuation of
the light pulse
paradigm. Notably, treated animals could freely switch directions of movement
and head
position from left to right and vice versa. In striking contrast with optical
HFS, optical
LFS (20 Hz) of the same afferent fibers worsened PD symptoms by driving
increased
ipsilateral rotational behavior (FIG. 8C), demonstrating that behavioral
effects seen do
not result from simply driving unilateral activity. Therefore, in contrast to
direct STN
cellular interventions, driving STN afferent fibers with HFS and LFS
differentially
modulated PD symptoms in a manner corresponding to frequencies of stimulation
linked
clinically to ameliorated or exacerbated PD symptoms.
A diverse array of fibers from widespread brain areas converge on the STN,
perhaps underlying the utility of the STN as a focal DBS target. Many of these
afferents
likely contribute together to the therapeutic effects, and it is unlikely that
a single source
of fibers completely accounts for the behavioral effects seen. However, we
explored these
afferents in greater detail to determine the general class of fibers that may
be
contributory.
Thy! ::ChR2 animals display ChR2 expression chiefly in excitatory projection
neurons. Indeed, the inhibitory markers GAD67 and GABA were not detectable in
Thyl ::ChR2 fibers within STN (Figure 9A, left), effectively ruling out
contributions from
the GABAergic pallidal projections (LGP/GPe). We also found no localization of
major
neuromodulatory markers (dopamine and acetylcholine) within the STN Thy!
::ChR2
fibers (FIG. 11C), ruling out dopaminergic SNr as a relevant fiber origin as
well. We next
explored possible sources of excitatory fibers, and found no expression of
ChR2-YFP in
the cell bodies of the excitatory parafascicular or pedunculopontine nuclei,
potential
contributors of excitatory fibers to the STN. Within neocortex of these mice,
however,
ChR2-YFP is expressed strongly in excitatory neurons that project to STN.
Since
pathologically strong connectivity between STN and primary motor cortex M1 has
been

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
33
suggested to underlie PD circuit dysfunction, we therefore explored M1 as a
possible
contributor.
We verified in Thyl ::ChR2 M1 the presence of strong and selective ChR2
expression largely restricted to layer V neurons and corresponding apical
dendrites but
not in cells within other layers (FIG. 9A, right). To probe the functional
connectivity
between these layer V projection neurons and STN in the PD animals, we
conducted a
separated-optrode experiment in anesthetized animals in which the fiberoptic
and
recording electrodes were placed in two different brain regions in Thyl ::ChR2
animals
(FIG. 9B). By driving M1 layer V projection neurons and simultaneously
recording in
both M1 and STN, we found that precise M1 stimulation of this kind potently
influenced
neural activity in the STN (FIG. 9C, FIG. 16C, D), and that M1 Layer V neurons
could be
antidromically recruited by optical stimulation in the STN (FIG. 15). While as
noted
above, many local afferents in the STN region, including from the ZI, are
likely to
underlie the complex therapeutic effects of DBS, functional influences between
M1 layer
V and STN could be a significant contributor. Indeed, we found that selective
M1 layer V
HFS optical stimulation sufficed to ameliorate PD symptoms in a similar manner
to STN
stimulation in an array of measures ranging from rotational behavior (FIG. 9D)
to head
position bias and locomotion (FIG. 9E, F). As with STN stimulation,
pathological
rotations and head position bias were reduced by optical HFS to Ml; in
contrast, while
not augmenting the pathology, optical 20 Hz (LFS) stimulation to M1 had no
therapeutic
effect (FIG. 9D, E, F), and even at the highest light intensities achievable
without
epileptogenesis, M1 LFS did not drive or modify rotational behavior, unlike M2
LFS
cortical stimulation that can elicit contralateral rotations. Finally,
increased functional
mobility with M1 HFS but not LFS was confirmed with quantification of
increased
distance and speed of locomotion in PD Thyl ::ChR2 mice; in the absence of
amphetamine, M1 HFS allowed the otherwise bradykinetic animals to move freely
without eliciting rotational behavior (FIG. 9F).
FIG. 4 shows direct optical inhibition of local STN neurons. (A) Cannula
placement, virus injection, and fiber depth were guided by recordings of the
STN, which
is surrounded by the silent zona incerta (ZI) and internal capsule (IC). (B)
Confocal
images of STN neurons expressing CaMKIIa::eNpHR-EYFP and labeled for
excitatory
neuron-specific CaMKIIa (right). (C) Continuous 561m illumination of the STN
expressing CaMKIIa::eNpHR-EYFP in anesthetized 6-0HDA rats reduced STN
activity;

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
34
representative optrode trace and amplitude spectrum shown. Mean spiking
frequency was
reduced from 29 3 Hz to 5 1 Hz (mean s.e.m., p < 0.001, Student's t-
test, n = 8
traces from different STN coordinates in 2 animals). (D) Amphetamine-induced
rotations
were not affected by stimulation of the STN in these animals (p> 0.05, n = 4
rats, t-test
with u = 0). The red arrow indicates direction of pathologic effects, while
the green arrow
indicates direction of therapeutic effects. The electrical control implanted
with a
stimulation electrode showed therapeutic effects with HFS (120-130 Hz, 60 tis
pulse
width, 130-200 A, p < 0.05, t-test with jt = 0). Percent change of -100%
indicates that
the rodent is fully corrected. Data in all figures are mean s.e.m. ns p> 0.05,
* p < 0.05,
** p < 0.01, *** p < 0.001.
FIG. 5 shows targeting astroglia within the STN. (A) Confocal images show STN
astrocytes expressing GFAP::ChR2-mCherry, costained with GFAP (right). (B)
473nm
illumination of the STN expressing GFAP::ChR2-mCherry in anesthetized 6-0HDA
rats.
Optrode recording revealed that continuous illumination inhibited STN activity
with 404
39 ms delay to onset and 770 + 82 ms delay to offset (n = 5 traces from
different STN
coordinates in 2 animals), while 50% duty cycle also inhibited spiking, with
delay to
onset of 520 40 ms and delay to offset of 880 29 ms (n = 3 traces from
different STN
coordinates in 2 animals) with p < 0.001. (C) Amphetamine-induced rotations
were not
affected by 50% duty cycle illumination in these animals (right, p> 0.05, n =
7 rats, t-test
with u = 0).
FIG. 6 shows optical depolarization of STN neurons at different frequencies.
(A)
Confocal images of STN neurons expressing CaMKIIa::ChR2-mCherry and labeled
for
the excitatory neuron specific CaMKIIa marker. (B) Optical HFS (120 Hz, 5 ms
pulse
width) of the STN expressing CaMKIIa::ChR2-mCherry in 6-0HDA rats recorded
with
the optrode connected to a 473nm laser diode (representative trace and
amplitude
spectrum shown). Frequency of spiking increased from 41 2 Hz to 85 2 Hz
(HFS vs.
pre, n = 5 traces: p <0.001, t-test, post, n = 3 traces; traces were sampled
from different
STN coordinates in 1 animal). (C) Amphetamine-induced rotations were not
affected by
high (left, 130 Hz, 5 ms, n = 5 rats) or low (middle, 20 Hz, 5 ms, n = 2 rats)
frequency
optical stimulation.
FIG. 7 shows quantification of the tissue volume recruited by optical
intervention.
(A) Intensity values for 473nm (blue) and 561m (yellow) light are shown for a
400 um
fiber as a function of depth in brain tissue. The dashed line at 1 mW/mm2 (30
mW light

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
source) indicates the minimum intensity required to activate channelrhodopsins
and
halorhodopsins. (B) Confocal images of STN neurons expressing CaMKIIa::ChR2-
mCheny and labeled for the immediate early gene product c-fos show robust
neuronal
activation produced by light stimulation in vivo. Arrowheads indicate c-fos
positive cells.
5 Freely moving rats expressing ChR2 in STN (same animals as in FIG. 6),
were stimulated
with 473nm light (20 Hz, 5 ms pulse width). (C) The STN volume that showed
strong c-
fos activation was estimated to be at least 0.7 mm3 (dashed lines indicate STN

boundaries); robust c-fos activation was observed medial-lateral (1.155 mm),
anterior-
posterior (0.800 mm), and dorsal-ventral (0.770 mm) on subthalamic slices
imaged by
10 confocal microscopy with DAPI counterstain.
FIG. 8 shows selective optical control of afferent fibers in the STN. (A)
Confocal
images of Thyl ::ChR2-EYFP expression in the STN and DAPI staining for nuclei
shows
selective expression in fibers and not cell bodies (right). (B) Optical HFS
(130 Hz, 5 ms
pulse width) of the STN region in an anesthetized Thyl ::ChR2-EYFP 6-0HDA
mouse
15 with 473nm light inhibited STN large-amplitude spikes (sample trace, top
left), while
inducing smaller-amplitude high-frequency oscillations (FIG. 13C, D; 14C).
Optical LFS
(20 Hz, 5 ms pulse width) produced reliable spiking at 20 Hz (bottom left).
While HFS
prevented bursting (top right, p <0.001, n = 3), LFS had no significant effect
on burst
frequency by 2 sample t-test (p 0.05, n = 3 traces) nor on spikes/burst
(bottom right, p>
20 0.05, n = 3 traces). (C) Optical HFS to STN in these animals (left, 100-
130 Hz, 5 ms, n =
5 mice) produced robust therapeutic effects, reducing ipsilateral rotations
and allowing
animals to freely switch directions. In contrast, optical LFS (second left, 20
Hz, 5 ms, n =
5 mice) exacerbated pathologic effects, causing increased ipsilateral
rotations. Both
effects were reversible (Post). Changes were significant by t-test with p. = 0
for both HFS
25 (p <0.001, n = 5 mice) and LFS (p <0.05, n = 5 mice) compared to
baseline (light off).
(F) Contralateral head position bias also showed robust correction with HFS by
2 sample
t-test (HFS vs. light off: p < 0.05; n = 2 mice), but not with LFS (LFS vs.
light off: p>
0.05, n = 2 mice).
FIG. 9 shows selective optical stimulation of layer V neurons in anterior
primary
30 motor cortex. (A) GAD67 and GABA staining showed no colocalization with
Thyl ::ChR2-EYFP in STN (left). Apical dendrites of layer V neurons can be
seen rising
to the pial surface (22, 23) (right). (B) Schematic for optical stimulation of
M1 with
simultaneous recording in STN of Thyl ::ChR2 mice. (C) Optical stimulation
(473nm) of

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
36
M1 and simultaneous recording in STN of anesthetized Thyl ::ChR2 mice. Optical
HFS
(130 Hz, 5 ms pulse width) of M1 modulated activity in both M1 and STN.
Optical LFS
(20 Hz, 5 ms) of M1 produced 20 Hz tonic firing in both M1 and STN. (D)
Optical HFS
(130 Hz, 5 ms pulse width) reduced amphetamine-induced ipsilateral rotations
in 6-
OHDA Thyl ::ChR2 mice (p < 0.01, n = 5 mice) in contrast to optical LFS (20
Hz, 5 ms
pulse width, p> 0.05, n = 4 mice); t-test with = 0. (E) Contralateral head
position bias
was corrected in HFS (HFS vs. light off: p < 0.001, n = 4 mice), while LFS had
little
effect (LFS vs. light off: p> 0.05, n = 3 mice); 2-sample t-test. (F) HFS but
not LFS to
M1 significantly increased path length (HFS, p < 0.01, n = 2 mice) and
climbing (p <
0.05, n = 3 mice); 2-sample t-test. Sample paths before, during, and after HFS
are shown
(100 seconds each, path lengths noted in cm).
According to a specific implementation, the following steps followed in
obtaining
the results discussed herein. To verify the phenotype of cells and measure c-
fos activity,
rodents were anaesthetized with 65 mg/kg sodium pentobarbital and
transcardially
perfused with ice-cold 4% paraformaldehyde (PFA) in PBS (pH 7.4). Brains were
fixed
overnight in 4% PFA and then equilibrated in 30% sucrose in PBS. 40 imi-thick
coronal
sections were cut on a freezing microtome and stored in cryoprotectant at 4 C
until
processed for immunohistochemistry. Free-floating sections were washed in PBS
and
then incubated for 30 min in 0.3% Triton X-100 (Tx100) and 3% normal donkey
serum
(NDS). Slices were incubated overnight with primary antibody in 0.01% Tx100
and 3%
NDS (rabbit anti-cfos 1:500, rabbit anti-GFAP 1:500, mouse anti-MAP2 1:500,
mouse
anti-GAD67 1:500, rabbit anti-GABA 1:200, mouse anti-vGlutl 1:500, mouse anti-
vGlut2 1:500, mouse anti-CaMKIIa 1:200, mouse anti-S10013 1:250, rabbit anti-
glutamate
1:200, chicken anti-tyrosine hydroxylase 1:500, and goat anti-choline
acetyltransferase
.. 1:200). Sections were then washed and incubated with secondary antibodies
(1:1000)
conjugated to FITC, Cy3 or Cy5 for 3 hrs at room temperature. Following a 20
min
incubation with DAPI (1:50,000) sections were washed and mounted on microscope

slides with PVA-DABCO.
Confocal fluorescence images were acquired on a scanning laser microscope
using
.. a 20X/0.70NA or a 40X/1.25NA oil immersion objective. To determine the
volume of
c-fos activation, serial stack images covering a depth of 20 ptrn through
multiple medial-
lateral, anterior-posterior and dorsal-ventral subthalamic sections were
acquired using
equivalent settings. The image analysis software calculated the number of c-
fos positive

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
37
cells per field by thresholding c-fos immunoreactivity above background levels
and using
the DAPI staining to delineate nuclei. To determine the rate of viral
transduction we
determined the percentage of CaMKIIa-immunoreactive neurons per 40X field that
were
also eNpHR-YFP positive in multiple serial stack images of subthalamic
sections. Large
field images of entire slices were collected on a Leica MZ16FA
stereomicroscope.
Lentiviral vectors carrying the genes used were constructed using cloning
techniques. The CaMKIIa::eNpHR construct was produced by PCR amplification of
the
eNpHR-EYFP construct previously published and cloned in-frame into the AgeI
and
EcoRI restriction sites of a lentivirus carrying the CaMKIIa promoter. The
CaMKIIa::ChR2 construct was produced by PCR amplification of the ChR2-mCherry
construct and was also cloned in-frame into the AgeI and EcoRI restriction
sites of a
lentivirus carrying the CaMKIIa promoter. The GFAP::ChR2 construct was
produced by
replacing the CaMKIIa promoter with the GFAP promoter in the CaMKIIa::ChR2-
mCherry construct using the AgeI and Pad restriction enzyme sites.
High titer lentivirus (>109 pfu/mL) was then produced via calcium phosphate co-

transfection of 293FT cells with the lentiviral vector, pCMVAR8.74 and pMD2.G
(S2).
24 h post-transfection, 293FT cells were switched to serum-free medium
containing 5
mM sodium butyrate; the supernatant was collected 16 h later and concentrated
by
ultracentrifugation at 50,000 x g with 20% sucrose cushion. The resulting
viral pellet was
resuspended in phosphate buffered saline at 1/1000th of the original volume.
To ensure that there would be no significant expression leak in non-targeted
cell
types, we employed a Cre-inducible AAV vector with a double-foxed inverted
open
reading frame (ORE), wherein the ChR2-EYFP sequence is present in the
antisense
orientation. Upon transduction, Cre-expressing cells invert the ChR2-EYFP ORE
in a
stable, irreversible fashion and thereby activate sustained ChR2-EYFP
expression under
control of the strong and constitutively active elongation factor la (EF-1a)
promoter
(Feng Zhang, unpublished results). To construct Cre-activated recombinant AAV
vectors,
the DNA cassette carrying two pairs of incompatible lox sites (loxP and
10x2722) was
synthesized and the ChR2-EYFP transgene was inserted between the loxP and
1ox2722
sites in the reverse orientation. The resulting double-foxed reverse ChR2-EYFP
cassette
was cloned into a modified version of the pAAV2-MCS vector carrying the EF-la
promoter and the Woodchuck hepatitis virus posttranscriptional regulatory
element
(WPRE) to enhance expression. The recombinant AAV vectors were serotyped with

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
38
AAV5 coat proteins and packaged by the viral vector core at the University of
North
Carolina. The final viral concentration was 2 x 1012 genome copies (gc) / mL.
Adult rats (female Fisher, 200-300 g) and mice (male and female, C57BL/6
background, 15-30 g) were the subjects of these experiments. Animal husbandry
and all
aspects of experimental manipulation of our animals were in strict accord with
guidelines
from the National Institute of Health and have been approved by members of the
Stanford
Institutional Animal Care and Use Committee. All surgeries were performed
under
aseptic conditions. Rodents were anaesthetized using 1.5% isoflurane (for
surgeries
longer than 1 hr) or i.p. injection (90 mg/kg ketamine and 5mg/kg xylazine for
rats; 80
mg/kg and 15-20 mg/kg, respectively, for mice). The top of the animal's head
was
shaved, cleaned with 70% ethanol and betadine and then placed in a
stereotactic
apparatus. Ophthalmic ointment was applied to prevent eye drying. A midline
scalp
incision was made and then small craniotomies were performed using a drill
mounted on
the stereotactic apparatus for the 6-0HDA injection in the medial forebrain
bundle (rat: -2
AP, 2 ML, -7.5 DV; mouse: -1.2 AP, 1.2 ML, -4.75 DV) and virus injection in
the STN
(rat: -3.6 mm AP, 2.5 mm ML; mouse: -1.9 mm AP, 1.7 mm ML).
For rodents that were injected with lentivirus in the STN, in vivo
extracellular
recording was used to accurately determine the location of the STN along the
dorsal-
ventral axis. The depth was around -7 mm in rats and -4 mm in mice. The
concentrated
lentivirus (described above) was delivered to the STN using a 10 1 syringe and
a thin 34
gauge metal needle; the injection volume and flow rate (3 sites within the STN
along the
dorsal-ventral axis; each injection was 0.6 pl at 0.1 1/min) was controlled
with an
injection pump. After the final injection, the needle was left in place for 10
additional
minutes and then slowly withdrawn.
6-0HDA was then used to lesion the substantia nigra and produce hemi-
Parkinsonian rodents. Desipramine (20mg/kg for rats; 10 mg/kg for mice;
noradrenergic
reuptake inhibitor to prevent damage to noradrenergic terminals) was
administered,
followed ¨30 minutes later by 6-0HDA (8 pg/4 pl for rats; 6 1g/2 p1 for mice)
with 0.1%
ascorbic acid (to prevent degradation of 6-0HDA) into the right medial
forebrain bundle
.. (rat: ¨2 AP , +2 ML, and -7.5 DV; mouse: -1.2 AP, +1.2 ML, and -4.75 DV).
The
perfusion for the 6-0HDA injection (rat: 4 pl, mouse 2 pl) was at the rate of
1.2 1/min
for 4 min, and the needle was left in situ for an additional 5 minutes.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
39
A fiber guide (rat: C312G, mouse: C313G) was beveled to form a sharp edge (to
more easily penetrate brain tissue and reduce tissue movement), and then
inserted through
the craniotomy to a depth of approximately 400 rim above the STN or the
anterior
primary motor cortex (mouse: 2 AP, 2 ML, 0.5 DV). One layer of adhesive cement
followed by cranioplastic cement was used to secure the fiber guide system to
the skull.
After 20 min, the scalp was sealed back using tissue adhesive. The animal was
kept on a
heating pad until it recovered from anesthesia. Buprenorphine (0.03 mg/kg) was
given
subcutaneously following the surgical procedure to minimize discomfort. A
dummy
cannula (rat: C312G, mouse: C313G) was inserted to keep the fiber guide
patent.
For electrical DBS control rodents, a stimulation electrode (MS303/3-B) was
implanted in the STN. The procedure above was followed for OHDA injection, in
vivo
extracellular recording was then used to determine the depth of the STN, and
the
stimulation electrode was inserted to that depth and secured using one layer
of adhesive
cement followed by cranioplastic cement. Tissue adhesive was used to reseal
the scalp,
the animal was kept on a heating pad until recovery from anesthesia and
buprenorphine
was given to minimize discomfort. A dust cap (303DC/1) was then used to cover
the
electrode contacts.
Simultaneous optical stimulation and electrical recording in a single region
in
living rodents was done as described previously using an optrode composed of
an
extracellular tungsten electrode (1 Mil, ¨125 rim) tightly attached to an
optical fiber
(-200 rim) with the tip of the electrode deeper (-0.4 mm) than the tip of the
fiber, to
ensure illumination of the recorded neurons. For stimulation and recording in
two distinct
regions, small craniotomies were created above both target regions, and a
fiber or optrode
was placed above one region through one craniotomy and a plain electrode or
optrode
was placed in the other region through a separate craniotomy (see FIG. 9B for
diagram).
Stimulation in the anterior motor cortex was achieved by placing the optical
fiber just
above the brain surface, activating layer 5 of the cortex; for STN
stimulation, the fiber
was 300 rim above the STN. The STN was identified using its highly stereotyped
firing
pattern and the fact that it is surrounded dorso-ventrally by silent regions.
The optical
fiber was coupled to a 473 nm or 561 nm laser diode (30 mW fiber output) from
CrystaLaser. Single unit recordings were done in rats anesthetized with 1.5%
isoflurane
and mice anesthetized with intraperitoneal injections of ketamine (80
mg/kg)/xylazine
(15-20 mg/kg) cocktail. pClamp 10 and a Digidata 1322A board were used to both
collect

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
data and generate light pulses through the fiber. The recorded signal was band
pass
filtered at 300Hz low/5 kHz high (1800 Microelectrode AC Amplifier). For
precise
placement of the fiber/electrode pair, stereotactic instrumentation was used.
For behavior, multimode optical fibers (NA 0.37; rat: 400 um core, BFL37-400;
5 .. mouse: 300 pm core, BFL37-300) were precisely cut to the optimal length
for
maximizing the volume of the STN receiving light. About one week before
behavior, an
extracellular recording electrode was used to determine the depth of the
dorsal border of
the STN from the tip of the cannula guide and fibers were cut to be 200-300
p.m shorter.
For anterior motor cortex stimulation, the fiber was placed above layer 5
(less than a
10 .. millimeter deep). To ensure stability of the fiber during testing in
moving animals, an
internal cannula adapter was glued to the stripped optical fiber. To insert
the fiber, rodents
were briefly placed under isoflurane and the fiber was inserted while the
animal was
recovering from anesthesia. The internal cannula adapter snapped onto the
cannula guide
and the bottom half of the plastic portion of a dummy cannula was also used to
ensure the
15 .. adapter remained connected to the top of the cannula guide.
For optical stimulation, the fiber was connected to a 473 nm or 561 nm laser
diode
(20 mW fiber output) through an FC/PC adapter. Laser output was controlled
using a
function generator (33220A) to vary the frequency, duty cycle, and intensity.
For
Thyl ::ChR2 animals, the average minimum intensity used to produce therapeutic
20 behavior was 10 mW. A custom aluminum rotating optical commutator was
used to
release torsion in the fiber caused by the animal's rotation.
Motor behavior was assessed using amphetamine-induced rotations, head position

bias, climbing, and track length. Animals were accepted for experimental
investigation
only if amphetamine reliably induced rotations in the ipsilateral direction
confirming a 6-
25 OHDA lesion of the substantia nigra. Before and after each stimulation
trial, a trial of
equal length with the light off was used as a control. Each of these trials
was about 3
minutes long making the entire off-on-off sequence 9 minutes long. For
amphetamine-
induced behavior, amphetamine (rat: 2 mg/kg; mouse: 2.6 mg/kg) was injected 30

minutes before behavioral measurements; the fiber was inserted into the
cannula and the
30 rodent placed in an opaque, non-reflective cylinder (rat: diameter 25
cm, height 61 cm;
mouse: diameter 20 cm, height 46 cm) 10 minutes before the behavioral
experiments.
Rotations ipsilateral to the 6-0HDA lesions (clockwise turns) were counted,
and
contralateral rotations were subtracted. The percentage change calculation
considered the

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
41
change in rotational bias relative to the period without stimulation. Head
position bias
was determined by counting the number of head tilts (>100 deviation left or
right of
midline) over time. Each time the rodent rose up and touched either paw to the
wall of the
cylinder was counted as an instance of climbing. Track length was measured
with
Viewer. After the completion of behavior experiments, cannula placement was
confirmed
by slicing.
For experiments where optical stimulation did not produce a change in the
rodent
behavior, we also collected path length and head position bias data while the
rodents were
under amphetamine. Continuous 561m illumination of the STN expressing
CaMKIIa::eNpHR-EYFP in 6-0HDA rats did not affect path length (cm/min; light
on vs.
light off: 757.05 163.11 vs. 785.74 157.56, p = 0.90, n = 4 rats; mean
s.e.m; 2-
sample t-test) or head position bias (% of time to the right; light on vs.
light off: 99.92
0.08 vs. 99.75 0.25, p = 0.56, n = 4 rats; mean s.e.m; 2-sample t-test).
Optical HFS
(120 Hz, 5 ms pulse width) or LFS (20 Hz, 5 ms pulse width) of the STN
expressing
CaMKIIa::ChR2-mCheffy in 6-0HDA rats did not affect path length (cm/min; HFS
vs.
light off: 803.82 129.04 vs. 851.95 166.20, p = 0.83, n = 5 rats; LFS vs.
light off:
847.15 141.95 vs. 779.11 104.01, p = 0.74, n= 2 rats; mean s.e.m; 2-
sample t-test)
or head position bias (% of time to the right; HFS vs. light off: 93.97 3.78
vs. 94.20
2.96, p = 0.96, n = 5 rats; LFS vs. light off: 98.50 1.50 vs. 98.50 0.50,
p = 1.00, n = 2
.. rats; mean 1 s.e.m; 2-sample t-test). 473nm illumination of the STN
expressing
GFAP::ChR2-mCheny in 6-0HDA rats also did not affect path length (cm/min;
light on
vs. light off: 1042.52 113.73 vs. 1025.47 113.63, p = 0.92, n = 4 rats;
mean s.e.m;
2-sample t-test) or head position bias (% of time to the right; light on vs.
light off: 98.16
0.98 vs. 98.98 0.65, p = 0.52, n = 4 rats; mean s.e.m; 2-sample t-test).
Light transmission measurements were conducted with blocks of brain tissue
prepared from two 300 g Fisher rats and immediately tested. Blocks of tissue 2
mm in
thickness were cut in 0 1 C sucrose solution using a vibratome. The tissue
was then
placed in a Petri dish containing the same sucrose solution over the
photodetector of a
power meter. The tip of a 200 mm diameter optical fiber coupled to a blue or
yellow diode
laser (473 nm or 561 nm, 30 mW fiber output) was mounted on a
micromanipulator. First,
the power was measured through the solution. Then, the tip of the fiber was
moved down
into the tissue in 100 m increments and the power was measured. When the
fiber
reached the Petri dish, the power measured was compared to the initial
measurement

CA 02722278 2010-10-22
WO 2009/131837
PCT/US2009/039949
42
through the solution to confirm the total power output through the fiber. The
percent
transmission fraction was then calculated as the ratio between the power
measured
through tissue and the power measured through solution. The power intensity
was then
calculated by considering the light intensity spread due to the conical shape
of the 30 mW
light output from a 400 pm fiber based on the fiber's numerical aperture of
0.37. The
fiber output was assumed to be uniform across the diameter of the cone.
Measurements
were made through grey matter in three blocks of brain tissue for each
wavelength with
one block each moving anterior-posterior in the thalamus and in the cortex and
dorsal-
ventral through the thalamus.
Threshold search in Clampfit was used for automated detection of spikes in the
multi-unit recording, which was then validated by visual inspection; the spike
waveforms
displayed by Clampfit were observed to check the quality of spike detection.
For traces
with multiple spike populations, thresholds were set to capture all the
spikes; during
bursting, it is likely that multiple neurons were recorded from
simultaneously. Bursts
were identified in Clampfit; any two consecutive spikes occurring in an
interval less than
300 ms were counted as belonging to the same burst and only bursts of at least
3 spikes
were included. To quantify the neural activity at different frequencies,
spectra for in vivo
extracellular recording traces were generated using a wavelet transform after
converting
the traces into binary spike trains. The trace was then converted into a
histogram with a
binwidth of 0.5 ms for each of the duration-matched pre-stimulation,
stimulation, and
post-stimulation epochs. The start and end times for each of the segments, as
well as the
number of spikes, are listed below.
Pre-Stimulation Light On Post-
Stimulation
Start End Spikes Start End Spikes Start End Spikes
Fig 4
32.5 72.5 413 102.5 142.5 84 175 215 435
(CaMKHa::eNpHR)
Fig 7
(CaNHCHa::ChR2, 5.28 10.4 238 15.38 20.5 477 22.48 27.6 235
IFS)
Fig 8
(Thyl::ChR2,HFS) 0 10.62 90 14.98 25.6 0 29.38 40 94
Fig 8 (Thyl::ChR2,
0 10.62 139 14.98 25.6 383 29.38 40 132
LFS)
Fig 9 (Thyl, HFS
0.8 4.8 55 15.46 19.46 28 26 30 30
Ml, M1 recd)
Fig 9 (Thyl, HFS 0.94 5.4 19 15 19.46 37 25.54 30
16

CA 02722278 2010-10-22
WO 2009/131837
PCT/US2009/039949
43
Ml, STN recd)
Fig 9 (Thyl, LFS
0 5.5 131 18.5 24 313 30.5 36 64
Ml, M1 reed)
Fig 9 (Thyl, LFS
0 5.5 50 18.5 24 115 30.5 36 39
Ml, STN recd)
Fig 13 (eNpfill,
32.5 72.5 263 102.5 142.5 84 175 215 248
small unit)
Fig 13 (eNpl1R,
32.5 72.5 114 102.5 142.5 0
175 215 145
large unit)
Table 1. The three segments of each power spectra were time matched; this
table
shows the segments of each trace (the start and end time in seconds), as well
as
the number of spikes detected during each period. Time intervals were chosen
to
reflect stationary states before, during, and after stimulation for each
trace, to
account for temporal delays in onset or offset of physiological effects.
The spike histograms were then convolved with a wavelet to measure the
amplitude
of the spectra at frequencies below 150 Hz over time. The average amplitude
over time
for each frequency was then plotted. The wavelet used is reproduced below.
g t) = e-t2 120-2 e-27ri ft
= 4/(3f).
For determining the change in activity of multiple frequency bands, amplitude
spectra for multiple duration-matched baseline and stimulation sweeps were
calculated as
described above. Mean amplitude within each frequency band was determined and
the
ratio of this value (stimulation/baseline) was calculated. Spike latencies of
the M1
response to optical stimulation of the STN were determined by measuring the
delay
between the first peaks in simultaneous optrode recordings of M1 and STN of a
Thyl ::ChR2-EYFP 6-0HDA mouse. 20 Hz, 5 ms pulse width of 473nm light was used
to
activate the STN.
FIG. 10 shows substantia nigra lesion and cannula track. Loss of nigral
dopaminergic cells following 6-0HDA administration in rat (A) and mouse (B):
coronal
slices (rat: AP -5.8; mouse AP -3) show decreased tyrosine hydroxylase levels
(red)
unilaterally in the substantia nigra pars compacta; SNc is outlined by white
brackets.
Insets below show higher resolution images of the lesioned (left) and
unlesioned (right)
sides of the substantia nigra. (C) Carmula track is visible in a coronal slice
showing
correct placement of the cannula above the STN area.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
44
FIG. 11 shows an additional histological characterization. (A) STN cells
expressing CaMKIIa::eNpHR-EYFP (green) label for the excitatory neuron
specific
glutamate marker (red). (B) STN cells expressing GFAP::ChR2-mCherry (red)
costain
with the astroglia-specific marker S10013 (green). In both (A) and (B) yellow
indicates
colocalization of the two markers. (C) Representative confocal images of TH
stain for
dopamine (top) and CHAT stain for acetylcholine (bottom) showed no
colocalization with
Thyl::ChR2-EYFP expression in the STN.
FIG. 12 shows additional behavioral results. (A) Continuous 473nm illumination

of the STN expressing GFAP::ChR2-mCherry in an anesthetized 6-0HDA mouse
completely inhibited STN activity. (B) and (C): Extension of mouse results.
(B)
Amphetamine-induced rotations were not affected by 50% duty cycle illumination
of the
GFAP::ChR2 expressing STN in 6-0HDA mice (n = 1 mouse and 2 sessions). (C)
Amphetamine-induced rotations were not affected by high (130 Hz, 5ms pulse
width, n =
1 mouse and 2 sessions) or low (20 Hz, 5ms, n = 1 mouse and 1 session)
frequency
optical stimulation in the CaMKIIa::ChR2 expressing STN in 6-0HDA mice. (D)
and
(C): Modulation of inhibitory neurons during behavior. Although mainly
excitatory, STN
has about 7-10% percent cells that stain for inhibitory neuronal markers, such
as
GAD65/67 and parvalbumin (Allen Brain Atlas). To obtain specific expression in
either
GAD67 or parvalbumin neurons we injected GAD67-Cre and parvalbumin-Cre mice
respectively (gift of Sylvia Arber) with a Cre-inducible adeno-associated
virus (AAV)
vector carrying ChR2-EYFP (Methods). Cre-dependent opsin expression was
observed in
the STN region, but behavior was unchanged with optical stimulation. (D)
Amphetamine-
induced rotations were not affected by high (130 Hz, 5ms, n = 2 mice and 4
sessions) or
low (20 Hz, 5ms, n = 1 mouse and 2 sessions) frequency optical stimulation in
6-0HDA
GAD67-Cre mice. (E) Amphetamine-induced rotations were not affected by high
(130
Hz, 5ms, n = 2 mice and 2 sessions) or low (20 Hz, 5ms, n = 2 mice and 2
sessions)
frequency optical stimulation in 6-0HDA parvalbumin-Cre mice.
FIG. 13 shows additional electrophysiological results. Isolation of large
amplitude (A) and small amplitude (B) units from the trace in FIG. 4C and
corresponding
power spectra. Red lines represent average waveforms for all superimposed
spikes that
occurred during 70s of baseline activity (n = 205 spikes for large amplitude
unit and n
428 spikes for small amplitude unit). Both small and large amplitude units
showed
decreased activity during light that returned to normal baseline levels after
stimulation.

CA 02722278 2010-10-22
WO 2009/131837 PCT/US2009/039949
(C) Response of STN to optical stimulation of STN in a Thyl ::ChR2-EYFP 6-0HDA

mouse at 90Hz. The STN is initially excited but activity is reduced in the
emergent
stationary state measured by loss of the large amplitude spikes evident during
the
baseline; nevertheless, significant low amplitude activity persists throughout
the
5 stimulation. (D) High-temporal resolution trace of the STN response to
optical
stimulation of STN in a Thyl ::ChR2-EYFP 6-0HDA mouse at 130Hz (see FIG. 5B
for
full trace). Again, the STN initially responds with a spike followed by low
amplitude
activity throughout stimulation. Changes in amplitude of the local circuit
responses can
reflect either altered recruited cell number or altered excitability of
recruited cellular
10 elements. While optrode recordings cannot report on the precise cell
types involved in
generating activity, by eliminating the electrical stimulation artifact these
recordings
provide a window into the amplitude and timing properties of local circuit
electrical
responses arising from local excitatory or inhibitory cell types and fibers in
the STN
region that could not be achieved with electrical stimulation.
15 FIG. 14 shows high-temporal resolution optrode traces. (A) Single unit
activity in
CaMKIIa::eNpHR-EYFP expressing STN with continuous 561 nm light illumination
in
an anesthetized 6-0HDA rat (corresponding to trace in FIG. 4C). (B) Neuronal
activity in
CaMKIIa::ChR2-mCherry expressing STN with high frequency optical stimulation
(120
Hz, 5 ms pulse width, 473 nm) in an anesthetized 6-0HDA rat (corresponding to
trace in
20 FIG. 3B). (C) and (D) Activity in the STN region in an anesthetized
Thyl::ChR2-EYFP
6-0HDA mouse in response to high (HFS, 130 Hz, 5 ms) and low (LFS, 20 Hz, 5
ms)
frequency optical stimulation using 473 nm light. Note the low amplitude of
activity in
the HFS trace (corresponding to trace in FIG. 5B).
FIG. 15 shows latency of M1 response to optical stimulation of STN. (A)
25 Response of M1 Layer 5 (L5) to optical stimulation of STN in the Thy 1
::ChR2-EYFP 6-
OHDA mouse at 20Hz, 5ms pulse width. (B) While stimulating STN with light,
simultaneous recordings of light-induced activity in the STN (top trace) and
Ml/L5
(bottom trace) revealed short latency differences between the first peaks
consistent with
antidromic spiking. (C) Individual latency differences between the first peak
in STN and
30 Ml/L5 for 16 stimulation bouts revealed minimal jitter (S.D. = 0.032 ms)
consistent with
antidromic spiking in the well-known Ml-STN projection.
FIG. 16 shows changes in frequency characteristics of neuronal activity
produced
by optical stimulation. (A) Activity in all frequency bands was reduced by
continuous

CA 02722278 2014-04-08
46
561m illumination of the STN expressing CaMKIla::eNpHR-EYFP in anesthetized 6-
OHDA rats (n = 5 sweeps). Frequency bands are defined as: delta 1-3 Hz; theta
4-8 Hz;
alpha 9-12 Hz; beta 13-30 Hz; gamma 31-80 Hz; high frequency (HF) 81-130 Hz.
(B)
Optical HFS (120 Hz, 5 ms pulse width) of the SIN expressing CaMKIla::ChR2-
mCherry in 6-0HDA rats reduced activity for frequencies between 4 and 80 Hz,
while
increasing activity in the IF band (n = 3). (C) Activity change in M1 (left n=
4) and
STN (right, n = 4) produced by optical ITS (130 Hz, 5 ms) stimulation of M1 in
6-
OHDA Thy! ::ChR2 mice. Delta activity in both M1 and SIN was reduced. (D)
Activity
change in M1 (left, n= 4) and STN (right, n= 4) produced by optical LFS (20
Hz, 5 ms)
stimulation of M1 in 6-0HDA Thyl ::ChR2 mice. Beta, gamma, and HF activity in
both
M1 and STN was increased. (B) Optical LFS (20 Hz, 5 ms) of the SIN in 6-0HDA
Thyl::ChR2 mice increased activity in the beta, gamma, and HF bands (n = 3).
(F) Spike
counts for duration-matched baseline and optical stimulation segments for each

experiment type. Optical stimulation of the STN expressing CaMKIIanGFAP-
mCherry
and optical HFS in 6-0HDA Thyl::ChR2 mice abolished spiking activity, reducing
activity across all frequencies to zero (not shown). Error bars are s.e.m.; t-
test with p. =
100 used for statistics, *p <0.05.
The various embodiments described above are provided by way of illustration
only and should not be construed to limit the invention. Based on the above
discussion
and illustrations, those skilled in the art will readily recognize that
various modifications
and changes may be made to the present invention without strictly following
the
exemplary embodiments and applications illustrated and described herein. For
instance,
such changes may include additional modifications to VChRl-based sequences.
Such
modifications and changes do not depart from the scope of the present
invention.

CA 02722278 2014-04-08
47
SEQUENCE TABLE
<210> 1
<211> 294
<212> PRT
<213> Artificial Sequence
<220>
<223> ChR1 Protein
<400> I
Met Ser Arg Arg Pro Trp Leu Leu Ala Leu Ala Leu Ala Val Ala Leu
1 5 10 15
Ala Ala Gly Ser Ala Gly Ala Ser Thr Gly Ser Asp Ala Thr Val Pro
20 25 30
Val Ala Thr Gin Asp Gly Pro Asp Tyr Val She His Arg Ala His Glu
35 40 45
Arg Met Leu Ser Ala Leu Cys Leu Met She Tyr Gly Tyr Gin Thr Trp
50 55 60
Lys Ser Thr Cys Gly Trp Glu Glu Ile Tyr Val Ala Thr Ile Glu Met
65 70 75 80
Ile Lys Phe Ile Ile Glu Tyr Phe His Glu She Asp Glu Pro Ala Val
85 90 95
Ile Tyr Ser Ser Asn Gly Asn Lys Thr Val Trp Leu Arg Tyr Ala Glu
100 105 110
Trp Leu Leu Thr Cys Arg Val Ile Leu Ile His Leu Ser Asn Leu Thr
115 120 125
Gly Leu Ala Asn Asp Tyr Asn Lys Arg Thr Met Gly Leu Leu Val Ser
130 135 140
Asp Ile Gly Thr Ile Val Trp Gly Thr Thr Ala Ala Leu Ser Lys Gly
145 150 155 160

CA 02722278 2014-04-08
,
48
Tyr Val Arg Val Ile Phe Phe Leu Met Gly Leu Cys Tyr Gly Ile Tyr
165 170 175
Thr Phe Phe Asn Ala Ala Lys Val Tyr Ile Glu Ala Tyr His Thr Val
180 185 190
Pro Lys Gly Ile Cys Arg Asp Leu Val Arg Tyr Leu Ala Trp Leu Tyr
195 200 205
Phe Cys Ser Trp Ala Met Phe Pro Val Leu Phe Leu Leu Gly Pro Glu
210 215 220
Gly Phe Gly His Ile Asn Gin Phe Asn Ser Ala Ile Ala His Ala Ile
225 230 235 240
Leu Asp Leu Ala Ser Lys Asn Ala Trp Ser Met Met Gly His Phe Leu
245 250 255
Arg Val Lys Ile His Glu His Ile Leu Leu Tyr Gly Asp Ile Arg Lys
260 265 270
Lys Gin Lys Val Asn Val Ala Gly Gin Glu Met Glu Val Glu Thr Met
275 280 285
Val His Glu Glu Asp Asp
290
<210> 2
<211> 305
<212> PRT
<213> Artificial Sequence
<220>
<223> ChR2 Protein
<400> 2
Met Asp Tyr Gly Gly Ala Leu Ser Ala Val Gly Arg Glu Leu Leu Phe
1 5 10 15
Val Thr Asn Pro Val Val Val Asn Gly Ser Val Leu Val Pro Glu Asp
20 25 30

CA 02722278 2014-04-08
õ .
49
Gin Cys Tyr Cys Ala Gly Trp Ile Glu Ser Arg Gly Thr Asn Gly Ala
35 40 45
Gin Thr Ala Ser Asn Val Leu Gin Trp Leu Ala Ala Gly Phe Ser Ile
50 55 60
Leu Leu Leu Met Phe Tyr Ala Tyr Gln Thr Trp Lys Ser Thr Cys Gly
65 70 75 80
Trp Glu Glu Ile Tyr Val Cys Ala Ile Glu Met Val Lys Val Ile Leu
85 90 95
Glu Phe Phe Phe Glu Phe Lys Asn Pro Ser Met Leu Tyr Leu Ala Thr
100 105 110
Gly His Arg Val Gin Trp Leu Arg Tyr Ala Glu Trp Leu Leu Thr Cys
115 120 125
Pro Val Ile Leu Ile His Leu Ser Asn Leu Thr Gly Leu Ser Asn Asp
130 135 140
Tyr Ser Arg Arg Thr Met Gly Leu Leu Val Ser Asp Ile Gly Thr Ile
145 150 155 160
Val Trp Gly Ala Thr Ser Ala Met Ala Thr Gly Tyr Val Lys Val Ile
165 170 175
Phe Phe Cys Leu Gly Leu Cys Tyr Gly Ala Asn Thr Phe Phe His Ala
180 185 190
Ala Lys Ala Tyr Ile Glu Gly Tyr His Thr Val Pro Lys Gly Arg Cys
195 200 205
Arg Gin Val Val Thr Gly Met Ala Trp Leu Phe Phe Val Ser Trp Gly
210 215 220
Met Phe Pro Ile Leu Phe Ile Leu Gly Pro Glu Gly Phe Gly Val Leu
225 230 235 240

CA 02722278 2014-04-08
. . .
. .
Ser Val Tyr Gly Ser Thr Val Gly His Thr Ile Ile Asp Leu Met Ser
245 250 255
Lys Asn Cys Trp Gly Leu Leu Gly His Tyr Leu Arg Val Leu Ile His
260 265 270
Glu His Ile Leu Ile His Gly Asp Ile Arg Lys Thr Thr Lys Leu Asn
275 280 285
Ile Gly Gly Thr Glu Ile Glu Val Glu Thr Leu Val Glu Asp Glu Ala
290 295 300
Glu
305
<210> 3
<211> 300
<212> PRT
<213> Artificial Sequence
<220>
<223> VChR1
<400> 3
Met Asp Tyr Pro Val Ala Arg Ser Leu Ile Val Arg Tyr Pro Thr Asp
1 5 10 15
Leu Gly Asn Gly Thr Val Cys Met Pro Arg Gly Gin Cys Tyr Cys Glu
20 25 30
Gly Trp Leu Arg Ser Arg Gly Thr Ser Ile Glu Lys Thr Ile Ala Ile
35 40 45
Thr Leu Gin Trp Val Val Phe Ala Leu Ser Val Ala Cys Leu Gly Trp
50 55 60
Tyr Ala Tyr Gin Ala Trp Arg Ala Thr Cys Gly Trp Glu Glu Val Tyr
65 70 75 80
Val Ala Leu Ile Glu Met Met Lys Ser Ile Ile Glu Ala Phe His Glu
85 90 95

CA 02722278 2014-04-08
" ' r
51
Phe Asp Ser Pro Ala Thr Leu Trp Leu Ser Ser Gly Asn Gly Val Val
100 105 110
Trp Met Arg Tyr Gly Glu Trp Leu Leu Thr Cys Pro Val Leu Leu Ile
115 120 125
His Leu Ser Asn Leu Thr Gly Leu Lys Asp Asp Tyr Ser Lys Arg Thr
130 135 140
Met Gly Leu Leu Val Ser Asp Val Gly Cys Ile Val Trp Gly Ala Thr
145 150 155 160
Ser Ala Met Cys Thr Gly Trp Thr Lys Ile Leu Phe Phe Leu Ile Ser
165 170 175
Leu Ser Tyr Gly Met Tyr Thr Tyr Phe His Ala Ala Lys Val Tyr Ile
180 185 190
Glu Ala Phe His Thr Val Pro Lys Gly Ile Cys Arg Glu Leu Val Arg
195 200 205
Val Met Ala Trp Thr Phe Phe Val Ala Trp Gly Met Phe Pro Val Leu
210 215 220
Phe Leu Leu Gly Thr Glu Gly Phe Gly His Ile Ser Pro Tyr Gly Ser
225 230 235 240
Ala Ile Gly His Ser Ile Leu Asp Leu Ile Ala Lys Asn Met Trp Gly
245 250 255
Val Leu Gly Asn Tyr Leu Arg Val Lys Ile His Glu His Ile Leu Leu
260 265 270
Tyr Gly Asp Ile Arg Lys Lys Gin Lys Ile Thr Ile Ala Gly Gin Glu
275 280 285
Met Glu Val Glu Thr Leu Val Ala Glu Glu Glu Asp
290 295 300

Representative Drawing

Sorry, the representative drawing for patent document number 2722278 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2020-03-31
(86) PCT Filing Date 2009-04-08
(87) PCT Publication Date 2009-10-29
(85) National Entry 2010-10-22
Examination Requested 2014-04-08
(45) Issued 2020-03-31
Deemed Expired 2020-08-31

Abandonment History

There is no abandonment history.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2010-10-22
Registration of a document - section 124 $100.00 2010-12-23
Maintenance Fee - Application - New Act 2 2011-04-08 $100.00 2011-03-29
Maintenance Fee - Application - New Act 3 2012-04-10 $100.00 2012-04-03
Maintenance Fee - Application - New Act 4 2013-04-08 $100.00 2013-04-03
Maintenance Fee - Application - New Act 5 2014-04-08 $200.00 2014-03-19
Request for Examination $800.00 2014-04-08
Maintenance Fee - Application - New Act 6 2015-04-08 $200.00 2015-03-25
Maintenance Fee - Application - New Act 7 2016-04-08 $200.00 2016-03-16
Maintenance Fee - Application - New Act 8 2017-04-10 $200.00 2017-03-28
Maintenance Fee - Application - New Act 9 2018-04-09 $200.00 2018-03-13
Maintenance Fee - Application - New Act 10 2019-04-08 $250.00 2019-03-26
Final Fee 2020-02-10 $300.00 2020-02-05
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Final Fee 2020-02-05 2 123
Cover Page 2020-03-09 1 33
Abstract 2010-10-22 1 58
Claims 2010-10-22 4 136
Drawings 2010-10-22 19 2,058
Description 2010-10-22 46 2,989
Cover Page 2011-01-19 1 35
Description 2014-04-08 53 3,147
Claims 2014-04-08 4 101
Description 2015-12-02 53 3,081
Claims 2015-12-02 4 106
Description 2016-12-02 53 3,083
Claims 2016-12-02 3 82
Examiner Requisition 2017-10-23 4 218
Amendment 2018-04-23 6 221
Claims 2018-04-23 4 129
Amendment 2018-05-02 3 103
Claims 2018-05-02 4 128
Prosecution-Amendment 2010-12-14 2 76
Maintenance Fee Payment 2018-03-13 1 33
Office Letter 2018-05-29 1 25
Examiner Requisition 2018-10-03 3 203
PCT 2010-10-22 13 544
Assignment 2010-10-22 3 82
Correspondence 2010-12-14 1 24
Correspondence 2010-12-23 2 62
Assignment 2010-12-23 7 341
Correspondence 2011-11-01 3 87
Assignment 2010-10-22 5 135
Maintenance Fee Payment 2019-03-26 1 61
Amendment 2019-04-02 7 257
Claims 2019-04-02 4 131
Correspondence 2012-05-17 4 117
Correspondence 2012-05-31 1 17
Correspondence 2012-05-31 1 20
Fees 2014-03-19 2 81
Prosecution-Amendment 2014-04-08 17 556
Prosecution-Amendment 2015-06-02 5 286
Amendment 2015-12-02 18 783
Correspondence 2015-02-17 3 252
Fees 2015-03-25 2 82
Assignment 2015-11-06 4 190
Examiner Requisition 2016-06-17 3 246
Amendment 2016-12-02 7 225
Amendment 2017-04-03 3 99

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :